US20180265570A1 - Albumin variants - Google Patents

Albumin variants Download PDF

Info

Publication number
US20180265570A1
US20180265570A1 US15/979,319 US201815979319A US2018265570A1 US 20180265570 A1 US20180265570 A1 US 20180265570A1 US 201815979319 A US201815979319 A US 201815979319A US 2018265570 A1 US2018265570 A1 US 2018265570A1
Authority
US
United States
Prior art keywords
albumin
hsa
variant
seq
fragment
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US15/979,319
Other versions
US10696732B2 (en
Inventor
Darrell Sleep
Andrew Plumridge
Jason Cameron
Inger Sandlie
Jan Terje Andersen
Esben Peter Friis
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Novozymes Biopharma DK AS
Albumedix Ltd
Original Assignee
Albumedix AS
Albumedix Ltd
Universitetet i Oslo
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to US15/979,319 priority Critical patent/US10696732B2/en
Application filed by Albumedix AS, Albumedix Ltd, Universitetet i Oslo filed Critical Albumedix AS
Assigned to ALBUMEDIX LTD reassignment ALBUMEDIX LTD ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NOVOZYMES BIOPHARMA DK A/S (BI-NAME ALBUMEDIX A/S)
Publication of US20180265570A1 publication Critical patent/US20180265570A1/en
Assigned to NOVOZYMES BIOPHARMA DK A/S reassignment NOVOZYMES BIOPHARMA DK A/S ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NOVOZYMES A/S, NOVOZYMES BIOPHARMA UK LIMITED, UNIVERSITY OF OSLO
Assigned to NOVOZYMES BIOPHARMA UK LIMITED reassignment NOVOZYMES BIOPHARMA UK LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SLEEP, DARRELL, CAMERON, JASON, PLUMRIDGE, ANDREW
Assigned to UNIVERSITY OF OSLO reassignment UNIVERSITY OF OSLO ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ANDERSEN, JAN TERJE, SANDLIE, INGER
Assigned to NOVOZYMES A/S reassignment NOVOZYMES A/S ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FRIIS, ESBEN PETER
Assigned to ALBUMEDIX A/S reassignment ALBUMEDIX A/S ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NOVOZYMES BIOPHARMA DK A/S
Priority to US16/882,019 priority patent/US20200385442A1/en
Publication of US10696732B2 publication Critical patent/US10696732B2/en
Application granted granted Critical
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/76Albumins
    • C07K14/765Serum albumin, e.g. HSA
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00ย -ย A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/20Fusion polypeptide containing a tag with affinity for a non-protein ligand
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/31Fusion polypeptide fusions, other than Fc, for prolonged plasma life, e.g. albumin

Definitions

  • the present invention relates to variants of albumin or fragments thereof or fusion polypeptides comprising variant albumin or fragments thereof having a change in half-life compared to the albumin, fragment thereof or fusion polypeptide comprising albumin or a fragment thereof.
  • Albumin is a protein naturally found in the blood plasma of mammals where it is the most abundant protein. It has important roles in maintaining the desired osmotic pressure of the blood and also in transport of various substances in the blood stream.
  • Albumins have been characterized from many species including human, pig, mouse, rat, rabbit and goat and they share a high degree of sequence and structural homology.
  • FcRn neonatal Fc receptor
  • HSA Human serum albumin
  • the plasma half-life of HSA has been found to be approximately 19 days.
  • a natural variant having lower plasma half-life has been identified (Peach, R. J. and Brennan, S. O., (1991) Biochim Biophys Acta. 1097:49-54) having the substitution D494N. This substitution generated an N-glycosylation site in this variant, which is not present in the wild-type albumin. It is not known whether the glycosylation or the amino acid change is responsible for the change in plasma half-life.
  • Albumin has a long plasma half-life and because of this property it has been suggested for use in drug delivery.
  • Albumin has been conjugated to pharmaceutically beneficial compounds (WO 2000/69902A), and it was found that the conjugateโ€”maintained the long plasma half-life of albumin.
  • the resulting plasma half-life of the conjugate was generally considerably longer than the plasma half-life of the beneficial therapeutic compound alone.
  • albumin has been fused to therapeutically beneficial peptides (WO 2001/79271 A and WO 2003/59934 A) with the typical result that the fusion has the activity of the therapeutically beneficial peptide and a considerably longer plasma half-life than the plasma half-life of the therapeutically beneficial peptides alone.
  • Galliano et al (1993) Biochim. Biophys. Acta 1225, 27-32 discloses a natural variant E505K.
  • Minchiotti et al. (1990) discloses a natural variant K536E.
  • Minchiotti et al (1987) Biochim. Biophys. Acta 916, 411-418 discloses a natural variant K574N.
  • Takahashi et al (1987) Proc. Natl. Acad. Sci. USA 84, 4413-4417 discloses a natural variant D550G. Carlson et al (1992).
  • Proc. Nat. Acad. Sci. USA 89, 8225-8229 discloses a natural variant D550A.
  • Albumin has the ability to bind a number of ligands and these become associated (associates) with albumin. This property has been utilized to extend the plasma half-life of drugs having the ability to noncovalently bind to albumin. This can also be achieved by binding a pharmaceutical beneficial compound, which has little or no albumin binding properties, to a moiety having albumin binding properties. See review article and reference therein, Kratz (2008). Journal of Controlled Release 132, 171-183.
  • Albumin is used in preparations of pharmaceutically beneficial compounds, in which such a preparation maybe for example, but not limited to, a nano particle or micro particle of albumin.
  • delivery of a pharmaceutically beneficial compound or mixture of compounds may benefit from alteration in the albumins affinity to its receptor where the beneficial compound has been shown to associate with albumin for the means of delivery.
  • the present invention provides variants of a parent albumin with improved properties compared to its parent.
  • the invention provides variants of a parent albumin having altered plasma half-life compare to its parent.
  • the present invention relates to isolated variants of albumin or fragments thereof, or fusion polypeptides comprising variant albumin or fragments thereof, of a parent albumin, comprising an alteration at one or more (several) positions corresponding to positions 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584 of the mature polypeptide of SEQ ID NO: 2, wherein the variant is not the variant consisting of SEQ ID NO: 2 with the substitution D494N, E501K, K541E, D550G,A, K573E or K574N.
  • the alteration at one or more position may independently be selected among substitutions, insertions and deletions, where substitution are preferred.
  • the present invention also relates to isolated polynucleotides encoding the variants; nucleic acid constructs, vectors, and host cells comprising the polynucleotides; and methods of producing the variants.
  • the present invention also relates to conjugates or associates comprising the variant albumin or fragment thereof according to the invention and a beneficial therapeutic moiety or to a fusion polypeptide comprising a variant albumin or fragment thereof of the invention and a fusion partner polypeptide.
  • the invention further relates to compositions comprising the variant albumin, fragment thereof, fusion polypeptide comprising variant albumin or fragment thereof or conjugates comprising the variant albumin or fragment thereof, according to the invention or associates comprising the variant albumin or fragment thereof, according to the invention.
  • the compositions are preferably pharmaceutical compositions.
  • the invention further relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a variant albumin, fragment thereof, fusion polypeptide comprising variant albumin or fragment thereof or conjugates comprising the variant albumin or fragment thereof, or associates comprising the variant albumin or fragment thereof, wherein said variant albumin, fragment thereof, fusion polypeptide comprising variant albumin or fragment thereof or conjugates comprising the variant albumin or fragment or associates of variant albumin or fragment thereof has altered plasma half-life compared to the corresponding plasma half-life of the HSA or fragment thereof, fusion polypeptide comprising HSA or fragment thereof or conjugates or associates of HSA or, fragment thereof, comprising HSA or fragment thereof.
  • FIG. 1 shows a restriction map of the expression plasmid pDB4082.
  • FIG. 2 shows a restriction map of the expression plasmid pDB2305
  • FIG. 3 shows a restriction map of the expression plasmid pDB4005
  • FIG. 5 shows ELISA binding of shFcRn-GST to human serum albumin (HSA) variants (100-0.045 โ‡ g/ml). Binding of WT, D494N, D494Q and D494A pH 6.0 and pH 7.4. Binding of WT, D494N, D494N/T496A and T496A at pH 6.0 and pH 7.4. Binding of WT, E495Q and E495A at pH 6.0 and pH 7.4.
  • HSA human serum albumin
  • FIG. 6 shows representative sensorgrams of binding of 0.2 โ‡ M of HSA variants to immobilized shFcRn ( โ‡ 4600 RU). WT, D494N, D494Q, D494A, D494N/T496A and T496A.
  • FIG. 7 shows representative sensorgrams of binding of 1 โ‡ M of HSA variants to immobilized shFcRn ( โ‡ 1400 RU). WT, D494N, D494Q, D494A, D494N/T496A and T496A.
  • FIG. 8 shows relative binding of the HSA variants compared to WT based on two independent SPR experiments as shown (A) FIG. 6 and (B) FIG. 7 .
  • FIG. 9 shows ELISA: (A) binding of shFcRn to albumins from human, donkey, bovine, sheep, goat and rabbit at pH 6.0. (B) binding of shFcRn to albumin from guinea pig, hamster, rat and chicken at pH 6.0. (C) binding of shFcRn to albumin from human, donkey, bovine, sheep, goat and rabbit at pH 7.4. (D) binding of shFcRn to albumin from guinea pig, hamster, rat and chicken at pH 7.4. (E) relative binding of the different albumins. Relative binding of human albumin to shFcRn is defined as 1.0. The ELISA values represent the mean of duplicates.
  • FIG. 10 shows SPR: Binding of shFcRn-GST to albumin from several species at pH 6.0 and pH 7.4. Representative sensorgrams showing binding of 5.0 โ‡ M of albumin from different species; (A) human, (B) donkey, (C) bovine, (D) goat, (E) sheep, (F) rabbit, (G) dog, (H) guinea pig, (I) hamster, (J) rat, (K) mouse and (L) chicken.
  • the albumin variants were injected over immobilized GST-tagged shFcRn ( โ‡ 2100 RU). Injections were performed at 25ยฐ C. at a rate of 40 โ‡ l/min.
  • FIG. 11 shows SPR sensorgrams of selected HSA mutants compared with wild-type HSA. 20 โ‡ M of (A) WT and P499A (B) WT and K500A, (C) WT and K536A, (D) WT and P537A and (E) WT and K538A and (F) WT and K537A were injected over immobilized shFcRn at pH 6.0 ( โ‡ 1500 RU)
  • FIG. 12 shows SPR sensorgrams of HSA mutants compared with WT HSA. 10 โ‡ M of (A) WT and K573A (B) WT and K573C, (C) WT and K573F, (D) WT and K573G and (E) WT and K573L and (F) WT and K573M, (G) WT and K573Q, (H) WT and K573R and (I) WT and K573T and (J) WT and K573V injected over immobilized shFcRn at pH 5.5 and pH7.4. Injections were performed at 25ยฐ C. at a flow rate of 80 โ‡ l/min.
  • FIG. 13 shows SPR sensorgrams of HSA mutants compared with wild-type HSA. 10 โ‡ M of (A) WT and K573D (B) WT and K573E, (C) WT and K573H, (D) WT and K5731 and (E) WT and K573N and (F) WT and K573P, (G) WT and K573S, (H) WT and K573* and (I) WT and K573W and (J) WT and K573Y injected over immobilized shFcRn at pH 5.5 and pH7.4. Injections were performed at 25ยฐ C. at a flow rate of 80 โ‡ l/min.
  • FIG. 14 shows SPR sensorgrams of HSA mutants compared with wild-type HSA. 20 โ‡ M of (A) WT and E492G+K538H+K541N+E542D (B) WT and E492T+N503K+K541A, (C) WT and E492P+N503K+K541G+E542P, (D) WT and E492H+E501P+N503H+E505D+T506S+T540S+K541E and (E) WT and A490D+E492T+V493L+E501P+N503D+A504E+E505K+T506F+K541D and (F) WT and E492G+V493P+K538H+K541N+E542D injected over immobilized shFcRn at pH 6.0. Injections were performed at 25ยฐ C. at a flow rate of 80 โ‡
  • FIG. 15 shows SPR sensorgrams of HSA mutants compared with wild-type HSA. Twenty โ‡ M of (A) WT, (B) H440Q, (C) H464Q and (D) H535Q injected over immobilized shFcRn at pH 6.0. Injections were performed at 25ยฐ C. at a flow rate of 80 โ‡ l/min.
  • FIG. 17 shows a restriction map of the expression plasmid pDB3017
  • FIG. 18 shows a restriction map of the expression plasmid pDB3021
  • FIG. 19 shows a restriction map of the expression plasmid pDB3056
  • FIG. 20 shows a restriction map of the expression plasmid pDB3165
  • FIG. 21 shows a restriction map of the expression plasmid pDB4172
  • FIG. 22 shows a restriction map of the expression plasmid pDB4267
  • FIG. 23 shows a restriction map of the expression plasmid pDB4285
  • FIG. 24 shows a GP-HPLC chromatogram of WT HSA and mutant K573P HRP conjugates for shFcRn analysis. Injections of 25 โ‡ L were made onto a TSK G3000SWXL column (Tosoh Bioscience) as described in materials and methods.
  • FIG. 25 shows SDS PAGE separation followed by both visual (A) and ultraviolet (B) detection of the Fluorescein conjugated albumin.
  • FIG. 26 shows shFcRn binding properties of HSA variants. 10 โ‡ M of WT rHA and E492T(A), WT rHA and D494N/E495Q/T496A(B), WT rHA and N503D(C), WT rHA and N503K(D), WT rHA and E492T/N503D(E), WT rHA and E495Q/T496A(F), WT rHA and K538H(G), WT rHA and E492D(H) injected over immobilised shFcRn at pH5.5
  • FIG. 27 shows shFcRn binding properties of HSA variants. 10 โ‡ M of WT rHA and K541A(I) and WT rHA and K541N(J) were injected over immobilised shFcRn at pH5.5.
  • FIG. 28 shows competitive binding of K573A and K573P measured by injecting shFcRn (100 nM) alone or pre-incubated with different amounts of HSA K573A and K573P over immobilized HSA ( โ‡ 2500 RU) at pH6.0
  • FIG. 29 shows competitive binding of HSA-FLAG variants measured by injecting shFcRn (100 nM) alone or together with different amounts of HSA-FLAG variants over immobilized HSA ( โ‡ 2500 RU) at pH6.0.
  • FIG. 30 shows competitive binding of HSA-IL1Ra variants measured by injecting shFcRn (100 nM) alone or together with different amounts of HSA-IL1Ra variants over immobilized HSA ( โ‡ 2500 RU) at pH6.0
  • FIG. 31 shows competitive binding of scFv-fused HSA variants measured by injecting shFcRn ( โ‡ 100 nM) alone or together with different amounts of (A) scFv-HSA-FLAG variants or (B) HSA-scFv-FLAG variants over immobilized HSA ( โ‡ 2500 RU) at pH6.0.
  • FIG. 32 shows binding of HSA, single, double and triple mutant variants to shFcRn.
  • Samples of 10 โ‡ M of each HSA variant were injected over immobilized shFcRn at pH 5.5 or pH 7.4.
  • the present invention relates to isolated variants of albumin or fragments thereof, or fusion polypeptides comprising variant albumin or fragments thereof, of a parent albumin, comprising an alteration at one or more (several) positions corresponding to positions 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584 of the mature polypeptide of SEQ ID NO: 2, wherein the variant is not the variant consisting of SEQ ID NO: 2 with the substitution D494N, E501K, K541E, D550G,A, K573E or K574N.
  • the alteration at one or more position may independently be selected among substitutions, insertions and deletions, where substitution are preferred.
  • variant means a polypeptide derived from a parent albumin by one or more alteration(s), i.e., a substitution, insertion, and/or deletion, at one or more (several) positions.
  • a substitution means a replacement of an amino acid occupying a position with a different amino acid;
  • a deletion means removal of an amino acid occupying a position; and
  • an insertion means adding 1 or more, preferably 1-3 amino acids immediately adjacent to an amino acid occupying a position.
  • Mutant means a polynucleotide encoding a variant.
  • Wild-Type Albumin means albumin having the same amino acid sequence as naturally found in an animal or in a human being.
  • parent or Parent albumin means an albumin to which an alteration is made by the hand of man to produce the albumin variants of the present invention.
  • the parent may be a naturally occurring (wild-type) polypeptide or an allele thereof, or even a variant thereof.
  • FcRn and shFcRn The term โ€œFcRnโ€ means the human neonatal Fc receptor (FcRn). shFcRn is a soluble recombinant form of FcRn.
  • smFcRn is a soluble recombinant form of the mouse neonatal Fc Receptor.
  • Isolated variant means a variant that is modified by the hand of man and separated completely or partially from at least one component with which it naturally occurs.
  • the variant is at least 1% pure, e.g., at least 5% pure, at least 10% pure, at least 20% pure, at least 40% pure, at least 60% pure, at least 80% pure, and at least 90% pure, as determined by SDS-PAGE or GP-HPLC.
  • substantially pure variant means a preparation that contains at most 10%, at most 8%, at most 6%, at most 5%, at most 4%, at most 3%, at most 2%, at most 1%, and at most 0.5% by weight of other polypeptide material with which it is natively or recombinantly associated.
  • the variant is at least 92% pure, e.g., at least 94% pure, at least 95% pure, at least 96% pure, at least 97% pure, at least 98% pure, at least 99%, at least 99.5% pure, and 100% pure by weight of the total polypeptide material present in the preparation.
  • the variants of the present invention are preferably in a substantially pure form. This can be accomplished, for example, by preparing the variant by well known recombinant methods and by purification methods.
  • Mature polypeptide means a polypeptide in its final form following translation and any post-translational modifications, such as N-terminal processing, C-terminal truncation, glycosylation, phosphorylation, etc.
  • the mature polypeptide is amino acids 1 to 585 of SEQ ID NO: 2, with the inclusion of any post-translational modifications.
  • Mature polypeptide coding sequence means a polynucleotide that encodes a mature albumin polypeptide.
  • the mature polypeptide coding sequence is nucleotides 1 to 1758 of SEQ ID NO: 1.
  • Sequence Identity The relatedness between two amino acid sequences or between two nucleotide sequences is described by the parameter โ€œsequence identityโ€.
  • the degree of sequence identity between two amino acid sequences is determined using the Needleman-Wunsch algorithm (Needleman and Wunsch, 1970 , J. Mol. Biol. 48: 443-453) as implemented in the Needle program of the EMBOSS package (EMBOSS: The European Molecular Biology Open Software Suite, Rice et al., 2000 , Trends Genet. 16: 276-277), preferably version 3.0.0 or later.
  • the optional parameters used are gap open penalty of 10, gap extension penalty of 0.5, and the EBLOSUM62 (EMBOSS version of BLOSUM62) substitution matrix.
  • the output of Needle labelled โ€œlongest identityโ€ is used as the percent identity and is calculated as follows:
  • the degree of sequence identity between two deoxyribonucleotide sequences is determined using the Needleman-Wunsch algorithm (Needleman and Wunsch, 1970, supra) as implemented in the Needle program of the EMBOSS package (EMBOSS: The European Molecular Biology Open Software Suite, Rice et al., 2000, supra), preferably version 3.0.0 or later.
  • the optional parameters used are gap open penalty of 10, gap extension penalty of 0.5, and the EDNAFULL (EMBOSS version of NCBI NUC4.4) substitution matrix.
  • the output of Needle labeled โ€œlongest identityโ€ (obtained using the -nobrief option) is used as the percent identity and is calculated as follows:
  • Fragment means a polypeptide having one or more (several) amino acids deleted from the amino and/or carboxyl terminus of an albumin and/or an internal region of albumin that has retained the ability to bind to FcRn. Fragments may consist of one uninterrupted sequence derived from HSA or it may comprise two or more sequences derived from HSA.
  • the fragments according to the invention have a size of more than approximately 20 amino acid residues, preferably more than 30 amino acid residues, more preferred more than 40 amino acid residues, more preferred more than 50 amino acid residues, more preferred more than 75 amino acid residues, more preferred more than 100 amino acid residues, more preferred more than 200 amino acid residues, more preferred more than 300 amino acid residues, even more preferred more than 400 amino acid residues and most preferred more than 500 amino acid residues.
  • allelic variant means any of two or more alternative forms of a gene occupying the same chromosomal locus. Allelic variation arises naturally through mutation, and may result in polymorphism within populations. Gene mutations can be silent (no change in the encoded polypeptide) or may encode polypeptides having altered amino acid sequences.
  • An allelic variant of a polypeptide is a polypeptide encoded by an allelic variant of a gene.
  • Coding sequence means a polynucleotide, which directly specifies the amino acid sequence of its translated polypeptide product.
  • the boundaries of the coding sequence are generally determined by an open reading frame, which usually begins with the ATG start codon or alternative start codons such as GTG and TTG and ends with a stop codon such as TAA, TAG, and TGA.
  • the coding sequence may be a DNA, cDNA, synthetic, or recombinant polynucleotide.
  • cDNA means a DNA molecule that can be prepared by reverse transcription from a mature, spliced, mRNA molecule obtained from a eukaryotic cell. cDNA lacks intron sequences that may be present in the corresponding genomic DNA.
  • the initial, primary RNA transcript is a precursor to mRNA that is processed through a series of steps, including splicing, before appearing as mature spliced mRNA.
  • nucleic acid construct means a nucleic acid molecule, either single- or double-stranded, which is isolated from a naturally occurring gene or is modified to contain segments of nucleic acids in a manner that would not otherwise exist in nature or which is synthetic.
  • nucleic acid construct is synonymous with the term โ€œexpression cassetteโ€ when the nucleic acid construct contains the control sequences required for expression of a coding sequence of the present invention.
  • control sequences means all components necessary for the expression of a polynucleotide encoding a variant of the present invention.
  • Each control sequence may be native or foreign to the polynucleotide encoding the variant or native or foreign to each other.
  • control sequences include, but are not limited to, a leader, polyadenylation sequence, propeptide sequence, promoter, signal peptide sequence, and transcription terminator.
  • the control sequences include a promoter, and transcriptional and translational stop signals.
  • the control sequences may be provided with linkers for the purpose of introducing specific restriction sites facilitating ligation of the control sequences within the coding region of the polynucleotide encoding a variant.
  • operably linked means a configuration in which a control sequence is placed at an appropriate position relative to the coding sequence of a polynucleotide such that the control sequence directs the expression of the coding sequence.
  • expression includes any step involved in the production of the variant including, but not limited to, transcription, post-transcriptional modification, translation, post-translational modification, and secretion.
  • Expression vector means a linear or circular DNA molecule that comprises a polynucleotide encoding a variant and is operably linked to additional nucleotides that provide for its expression.
  • host cell means any cell type that is susceptible to transformation, transfection, transduction, and the like with a nucleic acid construct or expression vector comprising a polynucleotide of the present invention.
  • host cell encompasses any progeny of a parent cell that is not identical to the parent cell due to mutations that occur during replication.
  • Plasma half-life is ideally determined in vivo in suitable individuals. However, since it is time consuming and expensive and there inevitable are ethical concerns connected with doing experiments in animals or man it is desirable to use an in vitro assay for determining whether plasma half-life is extended or reduced. It is known that the binding of albumin to its receptor FcRn is important for plasma half-life and the correlation between receptor binding and plasma half-life is that a higher affinity of albumin to its receptor leads to longer plasma half-life. Thus for the present invention a higher affinity of albumin to FcRn is considered indicative of an increased plasma half-life and a lower affinity of albumin to its receptor is considered indicative of a reduced plasma half-life.
  • a longer plasma half-life with respect to a variant albumin of the invention means that the variant has longer plasma half-life than the corresponding albumin having the same sequences except for the alteration(s) in positions corresponding to 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584 in SEQ ID NO: 2.
  • the mature polypeptide disclosed in SEQ ID NO: 2 is used to determine the corresponding amino acid residue in another albumin.
  • the amino acid sequence of another albumin is aligned with the mature polypeptide disclosed in SEQ ID NO: 2, and based on the alignment, the amino acid position number corresponding to any amino acid residue in the mature polypeptide disclosed in SEQ ID NO: 2 is determined using the Needleman-Wunsch algorithm (Needleman and Wunsch, 1970 , J. Mol. Biol. 48: 443-453) as implemented in the Needle program of the EMBOSS package (EMBOSS: The European Molecular Biology Open Software Suite, Rice et al., 2000 , Trends Genet. 16: 276-277), preferably version 3.0.0 or later.
  • EMBOSS European Molecular Biology Open Software Suite, Rice et al., 2000 , Trends Genet. 16: 276-277
  • proteins of known structure For proteins of known structure, several tools and resources are available for retrieving and generating structural alignments. For example the SCOP superfamilies of proteins have been structurally aligned, and those alignments are accessible and downloadable.
  • Two or more protein structures can be aligned using a variety of algorithms such as the distance alignment matrix (Holm and Sander, 1998 , Proteins 33: 88-96) or combinatorial extension (Shindyalov and Bourne, 1998 , Protein Engineering 11: 739-747), and implementations of these algorithms can additionally be utilized to query structure databases with a structure of interest in order to discover possible structural homologs (e.g., Holm and Park, 2000 , Bioinformatics 16: 566-567).
  • the distance alignment matrix Holm and Sander, 1998 , Proteins 33: 88-96
  • combinatorial extension Shindyalov and Bourne, 1998 , Protein Engineering 11: 739-747
  • albumin variants of the present invention the nomenclature described below is adapted for ease of reference.
  • the accepted IUPAC single letter or three letter amino acid abbreviation is employed.
  • substitutions For an amino acid substitution, the following nomenclature is used: Original amino acid, position, substituted amino acid. Accordingly, for example the substitution of threonine with alanine at position 226 is designated as โ€œThr226Alaโ€ or โ€œT226Aโ€. Multiple mutations are separated by addition marks (โ€œ+โ€), e.g., โ€œGly205Arg+Ser411Pheโ€ or โ€œG205R+S411Fโ€, representing substitutions at positions 205 and 411 of glycine (G) with arginine (R) and serine (S) with phenylalanine (F), respectively. The Figures also use (โ€œ/โ€), e.g., โ€œE492T/N503Dโ€ this should be viewed as interchangeable with (โ€œ+โ€).
  • Insertions For an amino acid insertion, the following nomenclature is used: Original amino acid, position, original amino acid, inserted amino acid. Accordingly the insertion of lysine after glycine at position 195 is designated โ€œGly195GlyLysโ€ or โ€œG195GKโ€. An insertion of multiple amino acids is designated [Original amino acid, position, original amino acid, inserted amino acid #1, inserted amino acid #2; etc.]. For example, the insertion of lysine and alanine after glycine at position 195 is indicated as โ€œGly195GlyLysAlaโ€ or โ€œG195GKAโ€.
  • the inserted amino acid residue(s) are numbered by the addition of lower case letters to the position number of the amino acid residue preceding the inserted amino acid residue(s).
  • the sequence would thus be:
  • variants comprising multiple alterations are separated by addition marks (โ€œ+โ€), e.g., โ€œArg170Tyr+Gly195Gluโ€ or โ€œR170Y+G195Eโ€ representing a substitution of tyrosine and glutamic acid for arginine and glycine at positions 170 and 195, respectively.
  • โ€œArg170Tyr,Gluโ€ represents a substitution of arginine with tyrosine or glutamic acid at position 170.
  • โ€œTyr167Gly,Ala+Arg170Gly,Alaโ€ designates the following variants: โ€œTyr167Gly+Arg170Glyโ€, โ€œTyr167Gly+Arg170Alaโ€, โ€œTyr167Ala+Arg170Glyโ€, and โ€œTyr167Ala+Arg170Alaโ€.
  • Albumins are proteins and constitute the most abundant protein in plasma in mammals and albumins from a long number of mammals have been characterized by biochemical methods and/or by sequence information.
  • albumins e.g., human serum albumin (HSA)
  • HSA human serum albumin
  • HSA is a preferred albumin according to the invention and is a protein consisting of 585 amino acid residues and has a molecular weight of 67 kDa. In its natural form it is not glycosylated.
  • the amino acid sequence of HSA is shown in SEQ ID NO: 2.
  • natural alleles may exist having essentially the same properties as HSA but having one or more amino acid changes compared to SEQ ID NO: 2, and the inventors also contemplate the use of such natural alleles as parent albumin according to the invention.
  • Albumins have generally a long plasma half-life of approximately 20 days or longer, e.g., HSA has a plasma half-life of 19 days. It is known that the long plasma half-life of HSA is mediated via interaction with its receptor FcRn, however, an understanding or knowledge of the exact mechanism behind the long half-life of HSA is not essential for the present invention.
  • albumin means a protein having the same, or very similar three dimensional structure as HSA and having a long plasma half-life.
  • albumin proteins can be mentioned human serum albumin, primate serum albumin, (such as chimpanzee serum albumin, gorilla serum albumin), rodent serum albumin (such as hamster serum albumin, guinea pig serum albumin, mouse albumin and rat serum albumin), bovine serum albumin, equine serum albumin, donkey serum albumin, rabbit serum albumin, goat serum albumin, sheep serum albumin, dog serum albumin, chicken serum albumin and pig serum albumin.
  • HSA as disclosed in SEQ ID NO: 2 or any naturally occurring allele thereof, is the preferred albumin according to the invention.
  • the parent albumin, a fragment thereof, or albumin part of a fusion polypeptide comprising albumin or a fragment thereof according to the invention has generally a sequence identity to the sequence of HSA shown in SEQ ID NO: 2 of at least 60%, preferably at least 70%, preferably at least 80%, preferably at least 85%, preferably at least 86%, preferably at least 87%, preferably at least 88%, preferably at least 89%, preferably at least 90%, preferably at least 91%, preferably at least 92%, preferably at least 93%, preferably at least 94%, preferably at least 95%, more preferred at least 96%, more preferred at least 97%, more preferred at least 98% and most preferred at least 99%.
  • the parent preferably comprises or consists of the amino acid sequence of SEQ ID NO: 2. In another aspect, the parent comprises or consists of the mature polypeptide of SEQ ID NO: 2.
  • the parent is an allelic variant of the mature polypeptide of SEQ ID NO: 2.
  • the parent is encoded by a polynucleotide that hybridizes under very low stringency conditions, low stringency conditions, medium stringency conditions, medium-high stringency conditions, high stringency conditions, or very high stringency conditions with (i) the mature polypeptide coding sequence of SEQ ID NO: 1, (ii) the mature polypeptide coding sequence of SEQ ID NO: 1, or (iii) the full-length complementary strand of (i) or (ii) (J. Sambrook, E. F. Fritsch, and T. Maniatis, 1989 , Molecular Cloning, A Laboratory Manual, 2d edition, Cold Spring Harbor, N.Y.).
  • the polynucleotide of SEQ ID NO: 1 or a subsequence thereof, as well as the amino acid sequence of SEQ ID NO: 2 or a fragment thereof, may be used to design nucleic acid probes to identify and clone DNA encoding a parent from strains of different genera or species according to methods well known in the art.
  • probes can be used for hybridization with the genomic or cDNA of the genus or species of interest, following standard Southern blotting procedures, in order to identify and isolate the corresponding gene therein.
  • Such probes can be considerably shorter than the entire sequence, but should be at least 14, e.g., at least 25, at least 35, or at least 70 nucleotides in length.
  • the nucleic acid probe is at least 100 nucleotides in length, e.g., at least 200 nucleotides, at least 300 nucleotides, at least 400 nucleotides, at least 500 nucleotides, at least 600 nucleotides, at least 700 nucleotides, at least 800 nucleotides, or at least 900 nucleotides in length.
  • Both DNA and RNA probes can be used.
  • the probes are typically labelled for detecting the corresponding gene (for example, with 32 P, 3 H, 35 S, biotin, or avidin). Such probes are encompassed by the present invention.
  • a genomic DNA or cDNA library prepared from such other organisms may be screened for DNA that hybridizes with the probes described above and encodes a parent.
  • Genomic or other DNA from such other organisms may be separated by agarose or polyacrylamide gel electrophoresis, or other separation techniques.
  • DNA from the libraries or the separated DNA may be transferred to and immobilized on nitrocellulose or other suitable carrier material.
  • the carrier material is used in a Southern blot.
  • hybridization indicates that the polynucleotide hybridizes to a labelled nucleotide probe corresponding to the polynucleotide shown in SEQ ID NO: 1, its complementary strand, or a subsequence thereof, under low to very high stringency conditions.
  • Molecules to which the probe hybridizes can be detected using, for example, X-ray film or any other detection means known in the art.
  • the nucleic acid probe is the mature polypeptide coding sequence of SEQ ID NO: 1. In another aspect, the nucleic acid probe is nucleotides 1 to 1785 of SEQ ID NO: 1. In another aspect, the nucleic acid probe is a polynucleotide that encodes the polypeptide of SEQ ID NO: 2 or a fragment thereof. In another aspect, the nucleic acid probe is SEQ ID NO: 1.
  • very low to very high stringency conditions are defined as prehybridization and hybridization at 42ยฐ C. in 5 โ‡ SSPE, 0.3% SDS, 200 micrograms/ml sheared and denatured salmon sperm DNA, and either 25% formamide for very low and low stringencies, 35% formamide for medium and medium-high stringencies, or 50% formamide for high and very high stringencies, following standard Southern blotting procedures for 12 to 24 hours optimally.
  • the carrier material is finally washed three times each for 15 minutes using 2 โ‡ SSC, 0.2% SDS at 45ยฐ C. (very low stringency), 50ยฐ C. (low stringency), 55ยฐ C. (medium stringency), 60ยฐ C. (medium-high stringency), 65ยฐ C. (high stringency), or 70ยฐ C. (very high stringency).
  • stringency conditions are defined as prehybridization and hybridization at about 5ยฐ C. to about 10ยฐ C. below the calculated T m using the calculation according to Bolton and McCarthy (1962 , Proc. Natl. Acad. Sci. USA 48: 1390) in 0.9 M NaCl, 0.09 M Tris-HCl pH 7.6, 6 mM EDTA, 0.5% NP-40, 1 โ‡ Denhardt's solution, 1 mM sodium pyrophosphate, 1 mM sodium monobasic phosphate, 0.1 mM ATP, and 0.2 mg of yeast RNA per ml following standard Southern blotting procedures for 12 to 24 hours optimally. The carrier material is finally washed once in 6 โ‡ SCC plus 0.1% SDS for 15 minutes and twice each for 15 minutes using 6 โ‡ SSC at 5ยฐ C. to 10ยฐ C. below the calculated T m .
  • the parent is encoded by a polynucleotide with a sequence identity to the mature polypeptide coding sequence of SEQ ID NO: 1 of at least 60%, e.g., at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%, which encodes a polypeptide which is able to function as an albumin.
  • the parent is encoded by a polynucleotide comprising or consisting of SEQ ID NO: 1.
  • the invention relates to a method for preparing a variant albumin, fragment thereof, or fusion polypeptide comprising variant albumin or a fragment thereof comprising the steps of:
  • the identification of one or more amino acid residue positions being important for the binding of albumin to FcRn, in albumin, fragment thereof or the albumin part of a fusion polypeptide can be done in several ways including, but not limited to, random mutagenesis followed by analysis of the generated mutants and comparison with the non-mutated parent molecule, and identification based on structural considerations optionally followed by generation of variants having the identified alterations and comparison with the non-mutated patent molecule.
  • a preferred method for identification of one or more amino acid residue positions to be changed to in order to prepare a variant HSA having an altered binding to FcRn compared with natural HSA comprises the following steps:
  • Step i) above may be done using the SPR assay described below.
  • SPR assay described below.
  • other methods may be used to identify non-human albumins having different binding properties to FcRn than HSA, and that the method is not dependent on how the non-human albumin, having different binding properties to FcRn, has been identified.
  • the identified non-human albumin has a stronger binding to FcRn than HSA.
  • non-human albumins having stronger binding to FcRn than HSA include donkey serum albumin, rabbit serum albumin, dog serum albumin, hamster serum albumin, guinea pig serum albumin, mouse serum albumin and rat serum albumin.
  • Step ii) may be accomplished by considering the structure of FcRn, HSA and the binding complex of these two. In the absence of an available structure of the binding complex it is possible to use a model where the HSA structure is docked into the structure of the FcRn structure and thereby identify amino acid residues of HSA interacting with FcRn.
  • the identified non-human albumin has a weaker binding to FcRn than HSA.
  • non-human albumins having weaker binding to FcRn than HSA include bovine serum albumin, goat serum albumin, sheep serum albumin and chicken serum albumin.
  • Step ii) may be accomplished by considering the structure of FcRn, HSA and the binding complex of these two. In absence of an available structure of the binding complex it is possible to use a model where the HSA structure is docked into the structure of the FcRn structure and thereby identify residues of HSA interacting with FcRn.
  • an amino acid residues of HSA interacting with FcRn is considered any amino acid residues of HSA being located less than 10A from an amino acid in the FcRn or any amino acid residue that is involved in a hydrogen bond, a salt bridge or a polar or nonpolar interaction with an amino acid residue that is located less than 10A from an amino acid in the FcRn.
  • the amino acid in HSA residues are located less than 10A from amino acids in the FcRn, more preferred less than 6A from amino acids in the FcRn and most preferred less than 3A from amino acids in the FcRn.
  • Step iii) and iv) can be done using techniques well known to the skilled person.
  • the present invention also relates to methods for obtaining a variant albumin or fragments thereof, or fusion polypeptides comprising the variant albumin or fragments thereof, or associates of variant albumin or fragment thereof comprising: (a) introducing into a parent albumin or fragments thereof, or fusion polypeptides comprising the parent albumin or fragments thereof an alteration at one or more (several) positions corresponding to positions 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584 of the mature polypeptide of SEQ ID NO: 2; and (b) recovering the variant albumin or fragments thereof, or fusion polypeptides comprising the variant albumin or fragments thereof.
  • the variants can be prepared by those skilled persons using any mutagenesis procedure known in the art, such as site-directed mutagenesis, synthetic gene construction, semi-synthetic gene construction, random mutagenesis, shuffling, etc.
  • Site-directed mutagenesis is a technique in which one or more (several) mutations are created at one or more defined sites in a polynucleotide encoding the parent.
  • Site-directed mutagenesis can be accomplished in vitro by PCR involving the use of oligonucleotide primers containing the desired mutation. Site-directed mutagenesis can also be performed in vitro by cassette mutagenesis involving the cleavage by a restriction enzyme at a site in the plasmid comprising a polynucleotide encoding the parent and subsequent ligation of an oligonucleotide containing the mutation in the polynucleotide. Usually the restriction enzyme that digests at the plasmid and the oligonucleotide is the same, permitting ligation of the plasmid and insert to one another. See, e.g., Scherer and Davis, 1979 , Proc. Natl. Acad. Sci. USA 76: 4949-4955; and Barton et al., 1990 , Nucleic Acids Res. 18: 7349-4966.
  • Site-directed mutagenesis can also be accomplished in vivo by methods known in the art. See, e.g., U.S. Patent Application Publication No. 2004/0171154; Storici et al., 2001 , Nature Biotechnol. 19: 773-776; Kren et al., 1998 , Nat. Med. 4: 285-290; and Calissano and Macino, 1996 , Fungal Genet. Newslett. 43: 15-16.
  • Any site-directed mutagenesis procedure can be used in the present invention.
  • Synthetic gene construction entails in vitro synthesis of a designed polynucleotide molecule to encode a polypeptide of interest. Gene synthesis can be performed utilizing a number of techniques, such as the multiplex microchip-based technology described by Tian et al. (2004 , Nature 432: 1050-1054) and similar technologies wherein oligionucleotides are synthesized and assembled upon photo-programmable microfluidic chips.
  • Single or multiple amino acid substitutions, deletions, and/or insertions can be made and tested using known methods of mutagenesis, recombination, and/or shuffling, followed by a relevant screening procedure, such as those disclosed by Reidhaar-Olson and Sauer, 1988 , Science 241: 53-57; Bowie and Sauer, 1989 , Proc. Natl. Acad. Sci. USA 86: 2152-2156; WO 95/17413; or WO 95/22625.
  • Other methods that can be used include error-prone PCR, phage display (e.g., Lowman et al., 1991 , Biochemistry 30: 10832-10837; U.S. Pat. No. 5,223,409; WO 92/06204) and region-directed mutagenesis (Derbyshire et al., 1986 , Gene 46: 145; Ner et al., 1988 , DNA 7: 127).
  • Mutagenesis/shuffling methods can be combined with high-throughput, automated screening methods to detect activity of cloned, mutagenized polypeptides expressed by host cells (Ness et al., 1999 , Nature Biotechnology 17: 893-896). Mutagenized DNA molecules that encode active polypeptides can be recovered from the host cells and rapidly sequenced using standard methods in the art. These methods allow the rapid determination of the importance of individual amino acid residues in a polypeptide.
  • Semi-synthetic gene construction is accomplished by combining aspects of synthetic gene construction, and/or site-directed mutagenesis, and/or random mutagenesis, and/or shuffling.
  • Semi-synthetic construction is typified by a process utilizing polynucleotide fragments that are synthesized, in combination with PCR techniques. Defined regions of genes may thus be synthesized de novo, while other regions may be amplified using site-specific mutagenic primers, while yet other regions may be subjected to error-prone PCR or non-error prone PCR amplification. Polynucleotide sub sequences may then be shuffled.
  • the present invention also provides variant albumins or fragments thereof, or fusion polypeptides comprising the variant albumin or fragments thereof, of a parent albumin, comprising an alteration at one or more (several) positions corresponding to positions 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584 in SEQ ID NO: 2, wherein each alteration is independently a substitution, insertion or deletion with the provision that the and the variant is not SEQ ID NO: 2 having the substitution D494N, E501K, K541E, D550G,A, K573E or K574N.
  • the variant albumin, a fragment thereof, or albumin part of a fusion polypeptide comprising variant albumin or a fragment thereof according to the invention has generally a sequence identity the sequence of HSA shown in SEQ ID NO: 2 of at least 60%, preferably at least 70%, preferably at least 80%, preferably at least 85%, preferably at least 90%, more preferred at least 95%, more preferred at least 96%, more preferred at least 97%, more preferred at least 98% and most preferred at least 99%.
  • the number of alterations in the variants of the present invention is 1-20, e.g., 1-10 and 1-5, such as 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 alterations.
  • the variant albumin, a fragment thereof or fusion polypeptide comprising the variant albumin or fragment thereof has altered plasma half-life compared with the corresponding parent albumin, fragment thereof, or fusion polypeptide comprising the variant albumin or fragment thereof.
  • the parent albumin is HSA and the variant albumin, a fragment thereof or fusion polypeptide comprising the variant albumin or fragment thereof has altered plasma half-life compared with the HSA, the corresponding fragment or fusion polypeptide comprising HSA or fragment thereof.
  • transgenic mouse having the natural mouse FcRn replaced with human FcRn has a higher serum albumin level than normal mouse; see (J Exp Med. (2003) 197(3):315-22).
  • human FcRn has a higher affinity to mouse serum albumin than mouse FcRn has to mouse serum albumin and, therefore, the observed increase in serum albumin in the transgenic mice corresponds with a higher affinity between serum albumin and its receptor, confirming the correlation between albumin binding to FcRn and plasma half-life.
  • variants of albumin that have little or no binding to FcRn have been shown to have reduced half-life in a mouse model, Kenanova et al (2009) J. Nucl. Med.; 50 (Supplement 2): 1582).
  • variant albumins having a KD that is lower than the KD for natural HSA is considered to have a higher plasma half-life than HSA and variant albumins having a KD that is higher than the KD for natural HSA is considered to have a lower plasma half-life than HSA.
  • the variants of albumin or fragments thereof or fusion polypeptides comprising albumin or fragments thereof comprise one or more alterations, such as substitutions, deletions or insertions at one or more (several) positions corresponding to the positions in HSA selected from the group consisting of 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584.
  • the substitution may be any substitution where the amino acid in the natural albumin sequence is substituted with a different amino acid selected among the remaining 19 natural occurring amino acids.
  • a variant comprises an alteration at one or more (several) positions corresponding to positions 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584 in SEQ ID NO: 2.
  • a variant comprises an alteration at two positions corresponding to any of 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584 in SEQ ID NO: 2.
  • a variant comprises an alteration at three positions corresponding to any of positions 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584 in SEQ ID NO: 2.
  • a variant comprises an alteration at each position corresponding to positions 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584 in SEQ ID NO: 2.
  • the variant comprises the substitution Q417A,H of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution H440Q of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution H464Q of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution A490D of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution E492G, T,P,H of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution V493P,L of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises the substitution D494N,Q,A,E,P of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises the substitution E495Q,A of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises the substitution T496A of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises the substitution P499A of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises the substitution K500E,G,D,A,S,C,P,H,F,N,W,T,M,Y,V,Q,L,I,R of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises the substitution E501A,P,Q of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution N503K,D,H of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution A504E of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution E505K, D of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution T506F, S of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution H510Q of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution H535Q of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises the substitution K536A of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises the substitution P537A of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises the substitution K538A,H of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises the substitution T540S of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises the substitution K541A,D,G,N,E of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises the substitution E542P,D of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises the substitution D550N of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises the substitution K573Y,W,P,H,F,V,I,T,N,S,G,M,C,A,E,Q,R,L,D of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises the substitution K574N of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises the substitution Q580K of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises the substitution L575F of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises the substitution A577T,E of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises the substitution A578R,S of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution S579C,T of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution Q580K of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution A581D of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution A582T of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution G584A of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 417.
  • the amino acid at a position corresponding to position 417 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Ala or His.
  • the variant comprises the substitution Q417A, H of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 440.
  • the amino acid at a position corresponding to position 440 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Ala.
  • the variant comprises the substitution H440Q of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 464.
  • the amino acid at a position corresponding to position 464 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Ala.
  • the variant comprises the substitution H464Q of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 490
  • the amino acid at a position corresponding to position 490 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val.
  • the variant comprises the substitution A490G of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 492.
  • the amino acid at a position corresponding to position 492 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Gly.
  • the variant comprises the substitution E492G of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 493.
  • the amino acid at a position corresponding to position 493 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Pro.
  • the variant comprises the substitution V493P of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 494.
  • the amino acid at a position corresponding to position 494 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Asn, Gin or Ala.
  • the variant comprises the substitution D494N,Q,A of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 495.
  • the amino acid at a position corresponding to position 495 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Gin or Ala.
  • the variant comprises the substitution E495Q or A of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 496.
  • the amino acid at a position corresponding to position 496 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Ala.
  • the variant comprises the substitution T496A of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 499.
  • the amino acid at a position corresponding to position 499 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Ala.
  • the variant comprises the substitution P499A of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 500.
  • the amino acid at a position corresponding to position 500 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Ala.
  • the variant comprises the substitution K500E,G,D,A,S,C,P,H,F,N,W,T,M,Y,V,Q,L,I,R of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 501.
  • the amino acid at a position corresponding to position 501 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Ala or Gln to reduce affinity and Pro to increase affinity.
  • the variant comprises the substitution E501A, Q, P of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 503.
  • the amino acid at a position corresponding to position 503 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Asp or Lys or His.
  • the variant comprises the substitution N503D, K, H of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 504.
  • the amino acid at a position corresponding to position 504 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val.
  • the variant comprises the substitution A504 of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 505.
  • the amino acid at a position corresponding to position 505 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val.
  • the variant comprises the substitution E505D of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 506.
  • the amino acid at a position corresponding to position 506 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val.
  • the variant comprises the substitution T506S,F of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 510.
  • the amino acid at a position corresponding to position 510 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Gln.
  • the variant comprises the substitution H510Q of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 535.
  • the amino acid at a position corresponding to position 535 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Gln.
  • the variant comprises the substitution H535Q of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 536.
  • the amino acid at a position corresponding to position 536 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Ala.
  • the variant comprises the substitution K536A of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 537.
  • the amino acid at a position corresponding to position 537 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Ala.
  • the variant comprises the substitution P537A of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 538.
  • the amino acid at a position corresponding to position 538 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Ala.
  • the variant comprises the substitution K538H, A of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 540.
  • the amino acid at a position corresponding to position 540 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val.
  • the variant comprises the substitution T540S of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 541.
  • the amino acid at a position corresponding to position 541 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Gly, Asp or Ala.
  • the variant comprises the substitution K541G, D A, N of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 542.
  • the amino acid at a position corresponding to position 542 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Asp or Pro.
  • the variant comprises the substitution E542D, P of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 550.
  • the amino acid at a position corresponding to position 550 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Asn to reduce affinity, preferably with Glu to increase affinity.
  • the variant comprises an alteration at a position corresponding to position 573.
  • the amino acid at a position corresponding to position 573 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Tyr, Trp, Pro, His. Phe, Val, Ile, Thr, Asn, Ser, Gly, Met, Cys, Ala, Glu, Gln, Arg, Leu, Asp.
  • the variant comprises the substitution K573Y,W,P,H,F,V,I,T,N,S,G,M,C,A,E,Q,R,L,D of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 574.
  • the amino acid at a position corresponding to position 574 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Asn.
  • the variant comprises the substitution K574N of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 575.
  • the amino acid at a position corresponding to position 575 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Phe.
  • the variant comprises the substitution L575F of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 577.
  • the amino acid at a position corresponding to position 577 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Thr or Glu.
  • the variant comprises the substitution A577TE of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 578.
  • the amino acid at a position corresponding to position 578 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Arg or Ser.
  • the variant comprises the substitution A578R,S of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 579.
  • the amino acid at a position corresponding to position 579 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Cys or Thr.
  • the variant comprises the substitution S579C,T of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 580.
  • the amino acid at a position corresponding to position 580 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Lys.
  • the variant comprises the substitution Q580K of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 581.
  • the amino acid at a position corresponding to position 581 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Asp.
  • the variant comprises the substitution A581D of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 582.
  • the amino acid at a position corresponding to position 582 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Thr.
  • the variant comprises the substitution A582T of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at a position corresponding to position 584.
  • the amino acid at a position corresponding to position 584 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Ala.
  • the variant comprises the substitution G584A of the mature polypeptide of SEQ ID NO: 2.
  • the variant comprises an alteration at positions corresponding to positions 494 and 496 in SEQ ID NO: 2, such as those described above.
  • the variant comprises alterations at positions corresponding to positions 492 and 493 in SEQ ID NO: 2, such as those described above.
  • the variant comprises alterations at positions corresponding to positions 494 and 417 in SEQ ID NO: 2, such as those described above.
  • the variant comprises alterations at positions corresponding to positions 492 and 503 in SEQ ID NO: 2, such as those described above.
  • the variant comprises alterations at positions corresponding to positions 492 and 573 in SEQ ID NO: 2, such as those described above.
  • the variant comprises alterations at positions corresponding to positions 492, 503, and 573 in SEQ ID NO: 2, such as those described above.
  • the variant albumin or fragments thereof, or fusion polypeptides comprising the variant albumin or fragments thereof according to the invention contains one substitution at a position corresponding to a position in HSA selected from the group consisting of 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584 in SEQ ID NO: 2 provided that the variant albumin is not the variant consisting of SEQ ID NO: 2 with the substitution D494N, E501K, K541E, D550G,A, K573E or K574N.
  • the variant albumin, fragment thereof or fusion polypeptides comprising variant albumin or a fragment thereof according to the invention may comprise additional substitutions, insertions or deletions at one or more
  • the variant albumin or fragments thereof, or fusion polypeptides comprising variant albumin or fragments thereof according to the invention contains two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen fifteen, sixteen, seventeen, eighteen, nineteen twenty or even more substitutions at positions corresponding to positions in HSA selected from the group consisting of 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584 of SEQ ID NO: 2.
  • the variant albumin or fragments thereof, or fusion polypeptides comprising variant albumin or fragments thereof according to the invention may comprise additional substitutions, insertions or deletions at positions corresponding to other positions in HSA.
  • variants of albumin or fragments thereof, or fusion polypeptides comprising variant albumin or a fragment thereof according to the invention have a plasma half-life that is longer than the plasma half-life of the parent albumin fragment thereof or fusion polypeptide comprising the parent albumin or a fragment thereof.
  • examples according to this embodiment include variants of albumin or fragments thereof, or fusion polypeptides comprising variant albumin or a fragment thereof comprising a substitution in the position corresponding to 492, 503, 542, 550, 573, 574, 580, 581, 582 or 584 in SEQ ID NO: 2.
  • Preferred substitutions according to this embodiment of the invention include the substitution of the amino acid residue in the position corresponding to 492 in SEQ ID NO: 2 with a G residue, substitution of the amino acid residue in the position corresponding to 503 in SEQ ID NO: 2 with a H or a K residue, substitution of the amino acid residue in the position corresponding to 550 in SEQ ID NO: 2 with an E residue, the substitution of the amino acid residue in a position corresponding to 573 in SEQ ID NO: 2 with an Y,W,P,H,F,V,I,T,N,S,G,M,C,A,E,Q,R,L or a D, the substitution of the amino acid residue in a position corresponding to 574 in SEQ ID NO: 2 with an N residue, or the substitution of the amino acid residue in the position corresponding to 580 in SEQ ID NO: 2 with an K residue.
  • Other preferred variants have a substitution in the position corresponding to 492 in SEQ ID NO: 2 with a G residue and a substitution in the position corresponding to 573 in SEQ ID NO: 2 with an A or a P residue.
  • Other preferred variant has a number of substitutions corresponding to position 492 in SEQ ID NO: 2 with an H residue in position 503 in SEQ ID NO: 2.
  • Other preferred variants have a substitution in the position corresponding to 492 in SEQ ID NO: 2 with a G residue and a substitution in the position corresponding to position 503 in SEQ ID NO: 2 corresponding to a H or a K and a substitution in position 573 in SEQ ID NO: 2 with an A or a P residue.
  • the variants of albumin or fragments thereof, or fusion polypeptides comprising variant albumin or fragments thereof according to the invention have a plasma half-life that is shorter than the plasma half-life of the parent albumin fragment thereof or fusion polypeptide comprising the parent albumin or a fragment thereof.
  • examples according to this embodiment include variants of albumin or fragments thereof, or fusion polypeptides comprising variant albumin or a fragment thereof comprising a substitution in the position corresponding to 417, 440, 494, 495, 496, 499, 500, 501, 536, 537, 538, 541, 494+496 or 492+493 in SEQ ID NO: 2.
  • Preferred substitutions include the substitutions corresponding to Q417A, H440Q, D494E+Q417H, D494N,Q,A, E495Q,A, T496A, D494N+T496A or, P499A, K500A, E501A, E501Q, K536A, P537A, K538A, K541G, K541A K541D or D550N in SEQ ID NO: 2.
  • variants of albumin or fragments thereof, or fusion polypeptides comprising variant albumin or a fragment thereof according to the invention have lost their ability to bind FcRn.
  • variants of albumin or fragments thereof, or fusion polypeptides comprising variant albumin or fragments thereof is considered to have lost the ability to bind FcRn if the measured resonance units for the variant in the SPR assay described below is less than 10% of the measured resonance units for the corresponding parent albumin or fragment thereof.
  • Examples according to this embodiment include variants of albumin or fragments thereof, or fusion polypeptides comprising variant albumin or fragments thereof comprising a substitution at a position corresponding to 464, 500, 510 or 535 in SEQ ID NO: 2.
  • Preferred substitutions include the substitutions corresponding to H464Q, K500A,P,C,S,A,D.G H510Q or H535Q in SEQ ID NO: 2.
  • the variant albumin or fragments thereof, or fusion polypeptides comprising variant albumin or fragments thereof according to the invention may contain additional substitutions, deletions or insertions in other positions of the molecules.
  • Such additional substitutions, deletions or insertions may be useful in order to alter other properties of the molecules such as but not limited to altered glycosylation; introduction of reactive groups of the surface such a thiol groups, removing/generating a carbamoylation site; etc.
  • Residues that might be altered in order to provide reactive residues on the surface and which advantageously could be applied to the present invention has been disclosed in the unpublished patent application WO 2010/092135 (Included by reference).
  • Particular preferred residues include the positions corresponding to positions in SEQ ID NO: 2.
  • cyste e
  • the present invention also relates to isolated polynucleotides that encode any of the variants of the present invention.
  • the present invention also relates to nucleic acid constructs comprising a polynucleotide encoding a variant of the present invention operably linked to one or more (several) control sequences that direct the expression of the coding sequence in a suitable host cell under conditions compatible with the control sequences.
  • a polynucleotide may be manipulated in a variety of ways to provide for expression of a variant. Manipulation of the polynucleotide prior to its insertion into a vector may be desirable or necessary depending on the expression vector. The techniques for modifying polynucleotides utilizing recombinant DNA methods are well known in the art.
  • the control sequence may be a promoter sequence, which is recognized by a host cell for expression of the polynucleotide.
  • the promoter sequence contains transcriptional control sequences that mediate the expression of the variant.
  • the promoter may be any nucleic acid sequence that shows transcriptional activity in the host cell including mutant, truncated, and hybrid promoters, and may be obtained from genes encoding extracellular or intracellular polypeptides either homologous or heterologous to the host cell.
  • useful promoters are obtained from the genes for Saccharomyces cerevisiae enolase (ENO-1), Saccharomyces cerevisiae protease A (PRA1), Saccharomyces cerevisiae protease B (PRB1), Saccharomyces cerevisiae translation elongation factor (TEF1), Saccharomyces cerevisiae translation elongation factor (TEF2), Saccharomyces cerevisiae galactokinase (GAL1), Saccharomyces cerevisiae alcohol dehydrogenase/glyceraldehyde-3-phosphate dehydrogenase (ADH1, ADH2/GAP), Saccharomyces cerevisiae triose phosphate isomerase (TPI), Saccharomyces cerevisiae metallothionein (CUP1), and Saccharomyces cerevisiae 3-phosphoglycerate kinase.
  • ENO-1 Saccharo
  • the control sequence may also be a suitable transcription terminator sequence, which is recognized by a host cell to terminate transcription.
  • the terminator sequence is operably linked to the 3โ€ฒ-terminus of the polynucleotide encoding the variant. Any terminator that is functional in the host cell may be used.
  • Preferred terminators for yeast host cells are obtained from the genes for Saccharomyces cerevisiae enolase, Saccharomyces cerevisiae cytochrome C (CYC1), Saccharomyces cerevisiae alcohol dehydrogenase (ADH1) and Saccharomyces cerevisiae glyceraldehyde-3-phosphate dehydrogenase.
  • CYC1 Saccharomyces cerevisiae cytochrome C
  • ADH1 Saccharomyces cerevisiae alcohol dehydrogenase
  • Saccharomyces cerevisiae glyceraldehyde-3-phosphate dehydrogenase Other useful terminators for yeast host cells are described by Romanos et al., 1992, supra.
  • the control sequence may also be a suitable leader sequence, a nontranslated region of an mRNA that is important for translation by the host cell.
  • the leader sequence is operably linked to the 5โ€ฒ-terminus of the polynucleotide encoding the variant. Any leader sequence that is functional in the host cell may be used.
  • Suitable leaders for yeast host cells are obtained from the genes for Saccharomyces cerevisiae enolase (ENO-1), Saccharomyces cerevisiae 3-phosphoglycerate kinase, Saccharomyces cerevisiae alpha-factor, and Saccharomyces cerevisiae alcohol dehydrogenase/glyceraldehyde-3-phosphate dehydrogenase (ADH2/GAP).
  • ENO-1 Saccharomyces cerevisiae enolase
  • Saccharomyces cerevisiae 3-phosphoglycerate kinase Saccharomyces cerevisiae alpha-factor
  • Saccharomyces cerevisiae alcohol dehydrogenase/glyceraldehyde-3-phosphate dehydrogenase ADH2/GAP
  • the control sequence may also be a polyadenylation sequence, a sequence operably linked to the 3โ€ฒ-terminus of the variant-encoding sequence and, when transcribed, is recognized by the host cell as a signal to add polyadenosine residues to transcribed mRNA. Any polyadenylation sequence that is functional in the host cell may be used.
  • yeast host cells Useful polyadenylation sequences for yeast host cells are described by Guo and Sherman, 1995 , Mol. Cellular Biol. 15: 5983-5990.
  • the control sequence may also be a signal peptide coding region that encodes a signal peptide linked to the N-terminus of a variant and directs the variant into the cell's secretory pathway.
  • the 5โ€ฒ-end of the coding sequence of the polynucleotide may inherently contain a signal peptide coding region naturally linked in translation reading frame with the segment of the coding region that encodes the variant.
  • the 5โ€ฒ-end of the coding sequence may contain a signal peptide coding region that is foreign to the coding sequence.
  • the foreign signal peptide coding region may be required where the coding sequence does not naturally contain a signal peptide coding region.
  • the foreign signal peptide coding region may simply replace the natural signal peptide coding region in order to enhance secretion of the variant.
  • any signal peptide coding region that directs the expressed variant into the secretory pathway of a host cell may be used.
  • Useful signal peptides for yeast host cells are obtained from the genes for Saccharomyces cerevisiae alpha-factor and Saccharomyces cerevisiae invertase. Other useful signal peptide coding sequences are described by Romanos et al., 1992, supra.
  • the propeptide region is positioned next to the N-terminus of the variant and the signal peptide region is positioned next to the N-terminus of the propeptide region.
  • the variants of the present invention can be prepared using techniques well known to the skilled person.
  • One convenient way is by cloning nucleic acid encoding the parent albumin or a fragment thereof or fusion polypeptide comprising albumin or a fragment thereof, modifying said nucleic acid to introduce the desired substitution(s) at one or more (several) positions corresponding to positions 417, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574 and 580 in SEQ ID NO: 2, where the variant is not the variant consisting of SEQ ID NO:2 with the substitution D494N, E501K, K541E, D550G,A, K573E or K574N., preparing a suitable genetic construct where the modified nucleic acid is placed in operative connection with suitable regulatory genetic elements, such as promoter, terminator,
  • the variant polypeptide of the invention may also be connected to a signal sequence in order to have the variant polypeptide secreted into the growth medium during culturing of the transformed host organism. It is generally advantageous to have the variant polypeptide secreted into the growth medium in order to ease recovery and purification.
  • Albumins have been successfully expressed as recombinant proteins in a range of hosts including fungi (including but not limited to Aspergillus (WO06066595), Kluyveromyces (Fleer 1991 , Bio/technology 9, 968-975), Pichia (Kobayashi 1998 Therapeutic Apheresis 2, 257-262) and Saccharomyces (Sleep 1990 , Bio/technology 8, 42-46)), bacteria (Pandjaitab 2000 , J. Allergy Clin. Immunol.
  • fungi including but not limited to Aspergillus (WO06066595), Kluyveromyces (Fleer 1991 , Bio/technology 9, 968-975), Pichia (Kobayashi 1998 Therapeutic Apheresis 2, 257-262) and Saccharomyces (Sleep 1990 , Bio/technology 8, 42-46)
  • bacteria Pandjaitab 2000 , J. Allergy Clin. Immunol.
  • the variant polypeptide of the invention is preferably produced recombinantly in a suitable host cell.
  • a suitable host cell In principle any host cell capable of producing a polypeptide in suitable amounts may be used and it is within the skills of the average practitioner to select a suitable host cell according to the invention.
  • a preferred host organism is yeast, preferably selected among Saccharomycacae, more preferred Saccharomyces cerevisiae.
  • variant polypeptides of the invention may be recovered and purified from the growth medium using a combination of known separation techniques such as filtration, centrifugation, chromatography, and affinity separation techniques etc. It is within the skills of the average practitioner to purify the variants of the invention using a particular combination of such known separation steps.
  • purification techniques that may be applied to the variants of the present invention can be mentioned the teaching of WO0044772.
  • the variant polypeptides of the invention may be used for delivering a therapeutically beneficial compound to an animal or a human individual in need thereof.
  • therapeutically beneficial compounds include, but are not limited, to labels and readily detectable compounds for use in diagnostics, such as various imaging techniques; pharmaceutical active compounds such as drugs, or specifically binding moieties such as antibodies.
  • the variants of the invention may even be connected to two or more different therapeutically beneficial compounds, e.g., an antibody and a drug, which gives the combined molecule the ability to bind specifically to a desired target and thereby provide a high concentration of the connected drug at that particular target.
  • the variants of albumin or fragments thereof according to the invention may also be fused with a non-albumin polypeptide fusion partner.
  • the fusion partner may in principle be any polypeptide but generally it is preferred that the fusion partner is a polypeptide having therapeutic or diagnostic properties.
  • Fusion polypeptides comprising albumin or fragments thereof are known in the art. It has been found that such fusion polypeptide comprising albumin or a fragment thereof and a fusion partner polypeptide have a longer plasma half-life compared to the unfused fusion partner polypeptide. According to the invention it is possible to alter the plasma half-life of the fusion polypeptides according to the invention compared to the corresponding fusion polypeptides of the prior art.
  • One or more therapeutic polypeptides may be fused to the N-terminus, the C-terminus of albumin, inserted into a loop in the albumin structure or any combination thereof. It may or it may not comprise linker sequences separating the various components of the fusion polypeptide.
  • WO 2001/79271 A and WO 2003/59934 A also contain examples of therapeutic polypeptides that may be fused to albumin or fragments thereof, and these examples apply also to the present invention.
  • the variants of albumin or fragments thereof according to the invention may be conjugated to a second molecule using techniques known within the art.
  • Said second molecule may comprise a diagnostic moiety, and in this embodiment the conjugate may be useful as a diagnostic tool such as in imaging; or the second molecule may be a therapeutic compound and in this embodiment the conjugate may be used for therapeutic purposes where the conjugate will have the therapeutic properties of the therapeutic compound as well as the long plasma half-life of the albumin.
  • Conjugates of albumin and a therapeutic molecule are known in the art and it has been verified that such conjugates have long plasma half-life compared with the non-conjugated, free therapeutic molecule as such.
  • the conjugates may conveniently be linked via a free thio group present on the surface of HSA (amino acid residue 34 of mature HSA) using well known chemistry.
  • the variant albumin or fragment thereof is conjugated to a beneficial therapeutic compound and the conjugate is used for treatment of a condition in a patient in need thereof, which condition is responsive to the particular selected therapeutic compound.
  • Techniques for conjugating such a therapeutically compound to the variant albumin or fragment thereof are known in the art.
  • WO 2009/019314 discloses examples of techniques suitable for conjugating a therapeutically compound to a polypeptide which techniques can also be applied to the present invention.
  • WO 2009/019314 discloses examples of compounds and moieties that may be conjugated to substituted transferrin and these examples may also be applied to the present invention. The teaching of WO 2009/019314 is included herein by reference.
  • HSA contains in its natural form one free thiol group that conveniently may be used for conjugation.
  • the variant albumin or fragment thereof may comprise further modifications provided to generate additional free thiol groups on the surface. This has the benefit that the payload of the variant albumin or fragment thereof is increased so that more than one molecule of the therapeutic compound can be conjugated to each molecule of variant albumin or fragment thereof, or two or more different therapeutic compounds may be conjugated to each molecule of variant albumin or fragment thereof, e.g., a compound having targeting properties such as an antibody specific for example a tumour; and a cytotoxic drug conjugated to the variant albumin or fragment thereof thereby creating a highly specific drug against a tumour.
  • teaching of particular residues that may be modified to provide for further free thiol groups on the surface can be found in copending patent application WO 2010/092135, which is incorporated by reference.
  • the variants of albumin or fragments thereof may further be used in form of โ€œassociatesโ€.
  • the term โ€œassociateโ€ is intended to mean a compound comprising a variant of albumin or a fragment thereof and another compound bound or associated to the variant albumin or fragment thereof by non-covalent binding.
  • an associate consisting variant albumin and a lipid associated to albumin by a hydrophobic interaction.
  • Such associates are known in the art and they may be prepared using well known techniques.
  • an associate comprising variant albumin and paclitaxel.
  • variant albumin or fragments thereof or fusion polypeptides comprising variant albumin or fragments thereof according to the invention have the benefit that their plasma half-life is altered compared to the parent albumin or fragments thereof or fusion polypeptides comprising parent albumin or fragments thereof.
  • This has the advantage that the plasma half-life of conjugates comprising variant albumin or a fragment thereof or fusion polypeptide comprising variant albumin or a fragment thereof, or an associate comprising variant albumin or a fragment thereof according to the invention can be selected in accordance with the particular therapeutic purpose.
  • a conjugate, associate or fusion polypeptide used for imaging purposes in animals or human beings where the imaging moiety has an very short half-life and a conjugate or a fusion polypeptide comprising HSA has a plasma half-life that is far longer than needed for the imaging purposes it would be advantageous to use a variant albumin or fragment thereof of the invention having a shorter plasma half-life than the parent albumin or fragment thereof, to provide conjugates of fusion polypeptides having a plasma half-life that is sufficiently long for the imaging purpose but sufficiently short to be cleared form the body of the particular patient on which it is applied.
  • an associate or fusion polypeptide comprising a therapeutic compound effective to treat or alleviate a particular condition in a patient in need for such a treatment it would be advantageous to use the variant albumin or fragment thereof having a longer plasma half-life than the parent albumin or fragment thereof, to provide associates or conjugates or fusion polypeptides having longer plasma half-lives which would have the benefit that the administration of the associate or conjugate or fusion polypeptide of the invention would be needed less frequently or reduced dose with less side affects compared to the situation where the parent albumin or associates thereof or fragment thereof was used.
  • compositions comprising the variant albumin, associates thereof or fragment thereof, variant albumin fragment or associates thereof or fusion polypeptide comprising variant albumin or fragment thereof according to the invention.
  • the compositions are preferably pharmaceutical compositions.
  • the composition may be prepared using techniques known in the area such as disclosed in recognized handbooks within the pharmaceutical field.
  • compositions comprise a variant albumin or a fragment thereof according to the invention and a compound comprising a pharmaceutically beneficial moiety and an albumin binding domain (ABD).
  • ABD means a site, moiety or domain capable of binding to circulating albumin in vivo and thereby conferring transport in the circulation of the ABD and any compound or moiety bound to said ABD.
  • ABD's are known in the art and have been shown to bind very tight to albumin so a compound comprising an ABD bound to albumin will to a certain extent behave as a single molecule.
  • the inventors have realized by using the variant albumin or fragment thereof according to the invention together with a compound comprising a pharmaceutically beneficial moiety and an ABD makes it possible to alter the plasma half-life of the compound comprising a pharmaceutically beneficial moiety and an ABD compared to the situation where said compound were injected as such in a patient having need thereof or administered in a formulation comprising natural albumin or a fragment thereof.
  • variant albumin or fragments thereof, conjugates comprising variant albumin or a fragment thereof or fusion polypeptide comprising variant albumin or a fragment thereof, or an associate comprising variant albumin or a fragment thereof according to the invention may also be incorporated into nano- or microparticles using techniques well known within the art.
  • a preferred method for preparing nano- or microparticles that may be applied to the variant albumins or fragments thereof according to the invention is disclosed in WO 2004/071536, which is incorporated herein by reference.
  • the present invention is also directed to the use of a variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, or a conjugate comprising a variant of albumin or a fragment thereof, or an associate comprising a variant of albumin or a fragment thereof for the manufacture of a pharmaceutical composition, where in the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, or a conjugate comprising a variant of albumin or a fragment thereof, or an associate comprising a variant of albumin or a fragment thereof has an altered plasma half-life compared with HSA or the corresponding fragment thereof or fusion polypeptide comprising HSA or fragment thereof or conjugate comprising HSA.
  • the corresponding fragment of HSA is intended to mean a fragment of HSA that aligns with and has same number of amino acids as the fragment of the variant albumin with which it is compared.
  • the corresponding fusion polypeptide comprising HSA or conjugate comprising HSA is intended to mean molecules having same size and amino acid sequence as the fusion polypeptide of conjugate comprising variant albumin, with which it is compared.
  • the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, or a conjugate comprising a variant of albumin or a fragment thereof has a plasma half-life that is higher than the plasma half-life of HSA or the corresponding fragment thereof or fusion polypeptide comprising HSA or fragment thereof.
  • this may be expressed as the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, or a conjugate comprising a variant of albumin or a fragment thereof has a KD to FcRn that is lower that the corresponding KD for HSA or the corresponding fragment thereof or fusion polypeptide comprising HSA or fragment thereof.
  • the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, or a conjugate comprising a variant of albumin or a fragment thereof is preferably the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, or a conjugate comprising a variant of albumin or a fragment thereof according to the invention.
  • Wells were coated with wild-type HSA or variants diluted in phosphate buffered saline (PBS) to stated concentrations, incubated overnight at 4 C and then blocked with 4% skimmed milk (Acumedia) for 1 hour at room temperature. The wells were then washed four times with PBS/0.005% TWEENยฎ 20 (PBS/T) pH 6.0 before glutathione-S-transferase (GST)-fused ( )shFcRn (0.5 โ‡ g/ml) as described in FEBS J. 2008 August; 275(16):4097-110.
  • PBS/T PBS/0.005% TWEENยฎ 20
  • GST glutathione-S-transferase
  • HRP horseradish peroxidase
  • PBS/T horseradish peroxidase pH 6.0
  • TMB substrate tetramethylbenzidine
  • HBS-EP buffer (0.01M HEPES, 0.15M NaCl, 3 mM EDTA, 0.005% surfactant P20
  • pH 7.4 0.05M HEPES, 0.15M NaCl, 3 mM EDTA, 0.005% surfactant P20
  • 1.0-0.5 โ‡ M of each HSA variant was injected over the surface at constant flow rate (40 โ‡ l/ml) at 25 C.
  • flow rate 40 โ‡ l/ml
  • HSA human serum albumin commercially available under the registered tradename RECOMBUMIN (available from Novozymes Biopharma UK Ltd, Nottingham UK) was used for the examples.
  • Serum albumin from other species The albumins were recombinant whereas stated, produced using sequences provided from publicly available databases. Or purchased from commercial suppliers.
  • FcRn Expression and purification of soluble Human (shFcRn) and Mouse (smFcRn) FcRn Methods for the generation of shFcRn and smFcRn expression plasmids, expression and purification of each heterodimer can be found in Berntzen et al. (2005) J. Immunol. Methods 298:93-104).
  • shFcRn FcRn heterodimer was produced by GeneArt AG (Germany). Sequences for the two sub units of the heterodimer can be found in SEQ ID NO: 3 and SEQ ID NO: 4.
  • the soluble receptor was expressed in HEK293 cells and purified from culture supernatant using Ni-HiTrap chromatography columns.
  • HSA mutant variants and HSA fusion variants were produced using several techniques. Standard molecular biology techniques were employed throughout such as described in Sambrook, J. and D. W. Russell, 2001. Molecular Cloning: a laboratory manual, 3rd ed. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.
  • Synthetic DNA NcoI/SacI fragments (859 bp) were generated by gene assembly (GeneArt AG, Germany) containing point mutations within the HSA-encoding gene (SEQ ID NO: 1) to introduce the desired amino acid substitution in the translated protein.
  • Table 2 details the codons used to introduce the amino acid substitutions into the HSA-encoding gene.
  • the nucleotide sequence of the synthetic fragment encoding unchanged amino acids was identical to that in pDB2243 (described in WO 00/44772).
  • the synthetic nucleotide fragments were ligated into NcoI/SacI-digested pDB2243 to produce plasmids pDB3876-pDB3886 (Table 1).
  • pDB3876-pDB3886 were each digested with NotI and PvuI, the DNA fragments were separated through a 0.7% (w/v) TAE gel, and 2992 bp fragments (NotI cassettes' including PRB1 promoter, DNA encoding the fusion leader (FL) sequence (disclosed in WO 2010/092135), nucleotide sequence encoding HSA and ADH1 terminator; see FIG. 1 ) were purified from the agarose gel using a Qiagen Gel Extraction Kit following the manufacturer's instructions.
  • NotI cassettes were ligated into a NotI/Shrimp Alkaline Phosphatase (Roche)-treated โ€œdisintegrationโ€ plasmid pSAC35, disclosed in EP-A-286 424 and described by Sleep, D., et al. (1991) Bio/Technology 9, 183-187. Ligation mixtures were used to transform chemically-competent E. coli DH5 โ‡ . Expression plasmids pDB3887-pDB3897, pSAC35-derivatives containing the โ€œNotI cassettesโ€, were identified using standard techniques.
  • Disintegration plasmids pDB3887-pDB3897 and pDB2244 were used to transform S. cerevisiae BXP10cir 0 (as previously described WO/2001/079480 as described below.
  • Oligonucleotide pairs xAP094 (SEQ ID NO: 5)/xAP095 (SEQ ID NO: 6) and xAP096 (SEQ ID NO: 7)/xAP097 (SEQ ID NO: 8) were used to generate two HSA variants (D494N+E495Q+T496A and E495Q+T496A, respectively).
  • Plasmid pDB3927 (disclosed in WO 2010/092135) was used as template DNA and the methodology recommended by the manufacturer of the kit was followed.
  • pDB3995 and pDB3996 were digested with BstEII/BsrBI and the linearised DNA molecules were purified using standard techniques.
  • BstEII/BsrBI digested DNA purified using a Qiagen PCR-Purification kit following the manufacturer's instructions, was mixed individually with 100 ng Acc65I/BamHI-digested pDB3936) (disclosed in WO 2010/092135) and used to directly transform S. cerevisiae BXP10cir 0 using the Sigma Yeast Transformation kit described below.
  • Plasmid pDB3927 (disclosed in WO 2010/092135) (containing an identical nucleotide sequence encoding HSA as in pDB2243) was manipulated to amino acid substitutions within the mature HSA protein.
  • Synthetic DNA fragments were generated (GeneArt AG, Germany or DNA2.0 Inc, USA) (NcoI/Bsu36I, AvrII/SphI or SacI/SphI fragments), containing point mutations within the HSA-encoding gene to introduce the desired amino acid substitution(s) into the translated protein sequence.
  • Table 2 details the codons used to introduce the amino acid substitutions into the HSA-encoding gene.
  • the nucleotide sequence of the synthetic fragment encoding unchanged amino acids i.e.
  • BamHI/SalI fragments containing point mutations in the nucleotide sequence encoding HSA were generated by gene assembly (DNA2.0 Inc, USA) and ligated into BamHI/SalI-digested pDB3964 (described in WO 2010/092135) to produce plasmids pDB3986-pDB3989 (Table 3).
  • the C-terminal string of amino acids from position 573-585 (KKLVAASQAALGL) (SEQ ID NO: 9) in HSA were mutated to those in macaque (PKFVAASQAALA) (SEQ ID NO: 10), mouse (PNLVTRCKDALA) (SEQ ID NO: 11), rabbit (PKLVESSKATLG) (SEQ ID NO: 12) and sheep (PKLVASTQAALA) (SEQ ID NO: 13) serum albumin.
  • the codons used to introduce each amino acid substitution are given in Table 2.
  • Synthetic DNA fragments (SacI/SphI) were generated (DNA2.0 Inc, USA) by gene assembly (the nucleotide sequence of the synthetic fragment encoding unchanged amino acids (i.e. wild type) was identical to that in pDB3927) and were sub-cloned into SacI/SphI-digested pDB3927 to produce plasmids pDB4114-4117 (Table 3).
  • Plasmids pDB3883 (Table 1), pDB4094 and pDB4095 (Table 3) were digested with NcoI/SacI and 857 bp fragments from each digest were purified before being ligated into NcoI/SacI-digested pDB4006 or pDB4110 (8.688 kb) (Table 3) to produce pDB4156-pDB4161.
  • Expression plasmids were generated in vivo (i.e. via homologous recombination in S. cerevisiae ; a technique referred to as gap repair or in vivo cloningโ€”see Orr-Weaver & Szostak. 1983 . Proc. Natl. Acad. Sci. USA. 80:4417-4421). Modified plasmids listed in Table 3 were digested with BstEII/BsrBI and the linearised DNA molecules were purified using standard techniques.
  • PCR was used to produce two permutation libraries in which the codons encoding amino acid 500 or 573 of mature HSA were changed (mutated) to alternative non-wild type amino acids and a termination codons (K5XXSTOP).
  • Mutagenic oligonucleotides (Table 4 and Table 5), were designed to amplify HSA-encoding DNA and incorporate the desired changes. That is, for the changes at position 500, pDB4082 ( FIG. 1 ) was used as a template DNA.
  • pDB4082 is a derivative of pDB2305 (disclosed in EP1788084) and was produced as follows.
  • pDB2305 FIG.
  • a synthetic DNA fragment (BsaI/SphI) was generated by gene assembly (DNA2.0 Inc, USA) (SEQ ID NO: 1) (containing 3โ€ฒ region of the PRB1 promoter, modified fusion leader sequence, nucleotide sequence encoding HSA and 5โ€ฒ region of the modified ADH1 terminator), and ligated into HindIII/SphI-digested pDB4005 ( FIG. 3 ) to produce pDB4082.
  • the HindIII site in PRB1 promoter site has been removed and a SacII site within the nucleotide sequence encoding HSA has been introduced.
  • the nucleotide sequence encoding HSA corresponding to that between the Sail/HindIII sites was generated using the New England Biolabs Phusion kit (Table 6) and oligonucleotides listed in Table 4.
  • Table 7 describes the PCR method employed.
  • the permutation library at amino acid position 573 in HSA was generated using pDB3927 as template DNA and involved amplifying the albumin-encoding DNA corresponding to that between the NcoI and Bsu36I sites using oligonucleotides detailed in Table 5.
  • each PCR-product was purified using a Qiagen PCR-clean up kit (according to the manufactures instructions), digested with SalI/HindIII (position 500 library) or NcoI/Bsu36I (position 573 library).
  • the digested DNAs were then purified using a Qiagen PCR-clean up kit and ligated into SalI/HindIII- or NcoI/Bsu36I-digested pDB4082 or pDB3927, respectively, replacing the equivalent native sequence. Ligations were transformed into E.
  • coli DH5 โ‡ subsequent plasmids isolated from transformants using a Qiagen miniprep kit (according to the manufacturer's instructions) and the correct constructs identified by restriction analysis.
  • the specific changes in each plasmid were confirmed by sequencing.
  • the resultants plasmids were used to generate expression plasmids and albumin fusion producing yeast by in vivo cloning as described above. That is, S. cerevisiae was transformed using the Sigma Yeast Transformation kit (described below), using a mixture of a 100 ng BstEII/BsrBI-digeste HSA variant containing plasmid and 100 ng Acc65I/BamHI digested pDB3936.
  • S. cerevisiae BXP10 cir 0 (as previously described WO/2001/079480) or Strain A cir 0 (described in WO/2005/061718) was streaked on to YEPD plates (1% (w/v) yeast extract, 2% (w/v) Bactopeptone, 2% (w/v) glucose), 1.5% agar) and allowed to grow for 4 days at 30ยฐ C. prior to transformation.
  • YEPD plates 1% (w/v) yeast extract, 2% (w/v) Bactopeptone, 2% (w/v) glucose), 1.5% agar
  • BMMD BMMD agar plates
  • the composition of BMMD is described by Sleep et al., (2001), Yeast, 18, 403. Plates were incubated at 30ยฐ C. for 4 days before individual colonies were patched on to fresh BMMD plates.
  • Yeast strain numbers are detailed in Table 1.
  • BMMD broth was inoculated with a heavy loop of each yeast patch and grown for 24 h at 30ยฐ C. with orbital shaking at 200 rpm.
  • Cells were harvested by centrifugation at 1900 โ‡ g for 5 min in a Sorval RT600 centrifuge, 15 mL supernatant was removed and replaced by trehalose 40% (w/v). The cells were resuspended and transferred to cyrovials (1 mL) for storage at โ‡ 80ยฐ C.
  • BMMD (recipe 0.17% (w/v) yeast nitrogen base without amino acid and ammonium sulphate (Difco), 37.8 mM ammonium sulphate, 29 mM citric acid, 142 mM disodium hydrogen orthophosphate dehydrate pH6.5, 2% (w/v) glucose) media (10 mL) was inoculated with each yeast strain and grown for 12 h at 30ยฐ C. with orbital shaking at 200 rpm. An aliquot of each starter culture (4 mL) was used to inoculate 2 โ‡ 200 mL BMMD media and grown for 36 h at 30ยฐ C. with orbital shaking at 200 rpm. Cells were harvested by filtration through 0.2 โ‡ m vacuum filter membranes (Stericup, Millipore) including a GF-D prefilter (Whatman) and the supernatant retained for purification.
  • Retained culture supernatant was concentrated using Tangential Flow Filtration using a Pall Filtron LV system fitted with a Omega 10KD (0.093 sq โ‡ m2) filter (LV CentramateTM cassette, Pall Filtron) with a transmembrane pressure of 20 psi and a recirculation rate of 180 mL โ‡ min โ‡ 1 .
  • a Pall Filtron LV system fitted with a Omega 10KD (0.093 sq โ‡ m2) filter (LV CentramateTM cassette, Pall Filtron) with a transmembrane pressure of 20 psi and a recirculation rate of 180 mL โ‡ min โ‡ 1 .
  • Sucrose was kept at growth-limiting concentrations by controlling the rate of feed to a set nominal growth rate.
  • the feed consisted of fermentation media containing 50% (w/v) sucrose, all essentially as described by Collins. (Collins, S. H., (1990) Production of secreted proteins in yeast, in: T. J. R. Harris (Ed.) Protein production by biotechnology, Elsevier, London, pp. 61-77).
  • Samples were chromatographed in 25 mM sodium phosphate, 100 mM sodium sulphate, 0.05% (w/v) sodium azide, pH 7.0 at 1 mL/min, Samples were quantified by UV detection at 280 nm, by peak area, relative to a recombinant human albumin standard of known concentration (10 mg/mL) and corrected for their relative extinction coefficients.
  • Albumin variants were purified from shake flask (either culture supernatant or concentrated culture supernatant) using a single chromatographic step using an albumin affinity matrix (AlbuPureTMโ€”ProMetic BioSciences, Inc.). Chromatography was performed at a constant linear velocity of 240 cm/h throughout. Culture supernatant was applied to a 6 cm bed height, 2.0 mL packed bed pre-equilibrated with 50 mM sodium acetate pH5.3. Following load the column was washed with 10 column volume (CV) of equilibration buffer, then 50 mM ammonium acetate pH8.0 (10CV).
  • CV column volume
  • Albumin-fusion variants were purified from shake flask culture supernatant using a single chromatographic step using an albumin affinity matrix (AlbuPureTMโ€”ProMetic BioSciences, Inc.). Chromatography was performed at a constant linear velocity of 240 cm/h throughout. Culture supernatant or concentrated culture supernatant was applied to a 6 cm bed height, 2.0 mL packed bed pre-equilibrated with 50 mM sodium acetate pH5.3. Following load the column was washed with 10 column volume (cv) equilibration buffer then 50 mM ammonium acetate pH8.0 (10cv).
  • albumin affinity matrix Albumin affinity matrix
  • Product was eluted with either 50 mM ammonium acetate 10 mM octanoate pH8.0, 50 mM Ammonium Acetate 30 mM Sodium Octanoate 200 mM Sodium Chloride pH7.0, 50 mM Ammonium Acetate 100 mM Sodium Octanoate pH9.0 or 200 mM Potassium thiocyanate.
  • the column was cleaned with 0.5M NaOH (3cv) and 20 mM NaOH (3.5cv).
  • Eluate fraction from each albumin variant-fusion were concentrated and diafiltered against 10 volumes of 25 mM Tris, 150 mM NaCl, 2 mM KCl, pH 7.4 (Vivaspin20 10,000 MWCO PES with optional diafiltration cups, Sartorius). Purified albumin-fusion variants were quantified by GP-HPLC as described above.
  • Albumin variants were purified from high cell density fed batch fermentation supernatants after separation by centrifugation, using a Sorvall RC 3C centrifuge (DuPont). Culture supernatant was chromatographed through an 11 cm bed height column 8.6 mL packed bed packed with a custom synthesised albumin affinity matrix (AlbuPureTMโ€”ProMetic BioSciences, Inc.) as described above. Product was eluted using elution buffers describe above at a flow rate of 120 cm/h. The eluate fraction(s) was analysed by GP-HPLC.
  • HSA Essentially fatty acid-free HSA (Sigma-Aldrich) was further purified by size exclusion chromatography as described in Andersen et al (2010). J. Biol. Chem. 285, (7), 4826-4836. Ten โ‡ M of monomeric HSA and rHA were analysed using SPR as described above and the data presented in FIG. 4 .
  • FIGS. 4A and 4B Direct comparison of HSA (blood derived) with recombinant human albumin (Recombumin) at the same concentration (10 โ‡ M) shows for both samples binding to immobilized shFcRn (pH6.0, pH7.4 respectively) was reversible and pH dependent.
  • the variants were analysed using ELISA at pH 6.0 and pH 7.4. Results are disclosed in FIG. 5 .
  • the ELISA values represent the mean of duplicates.
  • the variants were analysed using SPR analysis at pH 6.0 and pH 7.4. Results are disclosed for a representative number of variants in FIG. 6 using a concentration of the variants of 0.2 โ‡ M and in FIG. 7 using a concentration of the variants of 1 โ‡ M.
  • FIGS. 6 and 7 were normalized and the relative binding of variants at each concentration is shown in FIGS. 8 A and B respectively.
  • phosphate buffer (67 mM phosphate buffer, 0.15 M NaCl, 0.005% TWEENยฎ 20) at pH 6.0 or pH 7.4, or HBS-P buffer (0.01 M HEPES, 0.15 M NaCl, 0.005% surfactant P20) at pH 7.4 were used as running buffer or dilution buffer.
  • Kinetic measurements were performed using a low density immobilized surface (100-200 resonance units (RU)).
  • the KD's were generated using the BIAevaluation 4.1 software) A Langmuir 1:1 ligand model was used throughout. The kinetic values represent the average of triplicates. ND means: Not determined. NA means: Not acquired
  • animal albumin (either Sigma-Aldrich or Calbiochem) were further purified as described in Andersen et al (2010). J. Biol. Chem. 285, (7), 4826-4836.
  • the binding of donkey serum albumin, bovine serum albumin, goat serum albumin, sheep serum albumin, rabbit serum albumin, dog serum albumin, hamster serum albumin, guinea pig albumin, rat serum albumin and chicken serum albumin to shFcRn was determined using the techniques described in Materials and Methods.
  • the ELISA results are disclosed in FIG. 9 A-D and the relative bindings summarized in FIG. 9 E.
  • binding constants for variants according to the invention were determined according to the methods described in Materials and Methods.
  • the KD's were generated using the BIAevaluation 4.1 software) A Langmuir 1:1 ligand model was used throughout. The kinetic values represent the average of triplicates. ND means: Not determined.
  • Example 1 variants of HSA having the substitutions Q417A and D494E+Q417H were constructed.
  • the kinetic properties of these variants were tested using the methods in Materials and Methods and are shown in Table 12.
  • Example 1 variants of HSA having the substitutions P499A, K500A, K536A, P537A, K538A and K573A were constructed.
  • the receptor binding properties of these variants were tested as described in Materials and Methods. Results are shown in FIG. 11 .
  • variants P499A, K536A, P537A and K538A had a reduced binding affinity to shFcRn relative to HSA.
  • variant K500A had almost completely lost its ability to bind to shFcRn and K573A had an increased binding affinity to shFcRn both relative to HSA.
  • Example 1 variants of HSA having the substitutions E501A and E501Q were constructed.
  • the kinetic properties of these variants were tested as described in Materials and Methods.
  • Example 1 variants of HSA having a substitution at position 573 were constructed. All variants at position 573 were generated and the receptor binding properties of these variants were tested as described in Materials and Methods but with SPR analysis performed at pH5.5. Results are shown in the table 14 below and FIGS. 12 and 13 .
  • the variants K573F, K573H, K573P, K573W and K573Y have more than 10 fold lower KD to shFcRn than the parent HSA.
  • the variant K573STOP is a truncated albumin having a stop codon in position 573.
  • the sensorgram for the K573STOP variant show significantly reduced binding compare to the WT HSA and generated a high KD.
  • the increased affinity that we have shown for the variant K573E a natural variant characterized by Otagiri (2009). Biol. Pharm. Bull. 32(4) 527-534, is predicted to have increased half-life in vivo.
  • Example 1 variants of HSA having the substitutions E492G, E492G+N503H, N503H, D550E, E492G+N503K, E542P, H440Q, K541G, K541D, D550N E492G+K538H+K541N+E542D, E492T+N503K+K541A, E492P+N503K+K541G+E542P, E492H+E501P+N503H+E505D+T506S+T540S+K541E, A490D+E492T+V493L+E501P+N503D+A504E+E505K+T506F+K541D, E492G+V493P+K538H+K541N+E542D were constructed. The receptor binding properties of these variants were tested as described in Materials and Methods, and the results are shown in
  • FIG. 15 shows SPR sensorgrams of these variants interacting with shFcRn as described in Materials and Methods.
  • Example 1 variants of HSA having a substitution at position 500 were constructed. All variants at position 500 were generated and the receptor binding properties of these variants were tested.
  • Biacore X, Biacore X100 and Sensor Chip CM5 were used for all analyses, both supplied by G E Healthcare.
  • shFcRn produced by GeneArt AG (Germany) (diluted to 10 โ‡ g/mL in 10 mM sodium acetate pH5.0 (G E Healthcare)) was immobilised on flow cell 2 (FC2) to levels between 1600-2200 response units (RU) via standard amine coupling as per manufacturers instructions (G E Healthcare). A blank immobilisation was performed on flow cell 1 (FC1) for it to serve as a reference cell.
  • KD Albumin ka kd KD b Req c variant (10 3 /Ms) (10 โ‡ 3 /s) ( โ‡ M) ( โ‡ M) K500R 4.42 7.21 1.63 K500I 5.18 10.9 2.1 WT 4.24 9.2 2.2 a K500L 3.73 11.9 3.2 K500Q 1.07 3.4 3.2 K500V 3.29 11.0 3.3 K500Y 3.97 14.6 3.7 K500M 2.48 21.5 8.7 K500T 1.2 13.4 11.2 K500W 0.5 5.4 11.7 K500N 1.3 18.2 14 K500F 5.17 73.7 14.3 K500H 4 63.8 16 K500P ND ND ND 51* K500C 2.38 124 52 K500S ND ND โ‡ ND 70.2* K500A 2.61 208 79.9 K500D ND ND ND 83.3* K500G ND ND ND 95.4* K500E KD not calculable see FIG.
  • Plasmids containing expression cassettes for the production of scFv (vHvL) genetically-fused to HSA, at either the N- or C-terminus or both, were modified to allow the production of albumin fusions using in vivo cloning (describe above). That is, pDB3017 ( FIG. 17 ), pDB3021 ( FIG. 18 ), pDB3056 ( FIG. 19 ) were digested with NsiI/SpeI and NsiI fragments corresponding 9.511 kb, 9.569 kb and 8.795 kb, respectively, were purified using standard techniques. Purified NsiI fragments were self-ligated and used to transform chemically competent E. coli DH5 โ‡ to produce pDB4168, pDB4169 and pDB4170, respectively (Table 18).
  • pDB3165 (containing the bivalent fusion) ( FIG. 20 ) was digested with NotI and the expression cassette (4.506 kb fragment) was purified before being ligated into NotI-digested pDB3927 to produce pDB4172 ( FIG. 21 , Table 18).
  • Synthetic SalI/Bsu36I DNA fragments (269 bp), which contain point mutations within the albumin encoding nucleotide sequence to introduce amino acid substitutions corresponding to K500A, or D550N or K573P into the translated albumin protein sequence, were generated by gene assembly (GeneArt AG, Germany).
  • the SalI/Bsu36I fragments were individually ligated into SalI/Bsu36I-digested pDB4168-pDB4170 and pDB4172 and used to transform chemically competent E. coli DH5 โ‡ using standard techniques to generate plasmids pDB4265-pDB4276 (Table 18).
  • a DNA fragment was generated by PCR (using standard techniques), to introduce a K573A substitution in the translated albumin protein sequence.
  • PCR was performed using the New England Biolabs Phusion kit using pDB4267 ( FIG. 22 ) as template DNA and oligonucleotides xAP238 (SEQ ID NO: 53) and xAP239 (SEQ ID NO: 54):
  • Table 19 describes PCR cycling.
  • PCR-product was purified, digested with SalI/Bsu36I, and the fragment (269 bp) isolated was ligated into SalI/Bsu36I-digested pDB4168-pDB4170 and pDB4172 and used to transform chemically competent E. coli DH5 โ‡ .
  • Resulting plasmids (pDB4277-pDB4280) are listed in Table 18.
  • the nucleotide sequence encoding the FLAG tag was removed from plasmids pDB4168 and pDB4268-4270 (plasmids for the expression of scFv N-terminally fused to HSA and HSA muteins K500A, D550N and K573P, respectively.
  • pDB4168 and pDB4268-4270 (Table 18) were digested with Bsu36I/SphI to remove a 231 bp product comprising 3โ€ฒ region of HSA-encoding gene, nucleotide sequence encoding FLAG tag and 5โ€ฒ region of ADH1 terminator.
  • a Bsu36I/SphI fragment (207 bp), comprising 3โ€ฒ region of HSA-encoding gene and 5โ€ฒ region of mADH1 terminator (SEQ ID1) from pDB4181 was ligated into Bsu36I/SphI-digested pDB4168 and pDB4268-pDB4270 using standard techniques. Ligation mixtures were used to transform chemically competent E. coli DH5 โ‡ using standard techniques to generate plasmids pDB4281-pDB4284 (Table 18)
  • pDB4265-pDB4284 were digested with BstEII/BsrBI and the linearised DNA molecules were purified using standard techniques.
  • One hundred ng BstEII/BsrBI DNA samples were mixed with 100 ng Acc65I/BamHI-digested pDB3936 and used to transform S. cerevisiae BXP10cir 0 using the Sigma Yeast Transformation kit described below.
  • the expression plasmid was generated in the yeast by homologous recombination (in vivo cloning) between the albumin-fusion containing plasmid (pDB4265-pDB4280) (Table 18) and pDB3936.
  • Plasmids pDB3017, pDB3021, pDB3056 and pDB3165 were used to transform S. cerevisiae Strain Acir 0 (described in WO/2005/061718) using the Sigma Yeast Transformation kit described below.
  • the nucleotide sequence encoding human IL-1RA (interleukin-1 receptor antagonist) (accession number: CAA59087) could be synthetically generated by gene assembly.
  • the nucleotide sequence of the 708 bp synthetic fragment (Bsu36I/SphI fragment) is given in SEQ ID NO: 55 and includes the 3โ€ฒ region of the gene encoding HSA, the nucleotide sequence encoding a GS linker, the nucleotide sequence encoding human IL-1RA (N84Q to abolish the N-linked glycosylation motif) and the 5โ€ฒ region of the ADH1 terminator.
  • the synthetic DNA fragment could be ligated into Bsu36I/SphI-digested pDB3927 to produce pDB2588.
  • Plasmids containing the expression cassettes for the production of IL-1RA genetically fused to the C-terminus of HSA and the HSA variants K500A, D550N, K573A and K573P were prepared as follows.
  • pDB2588 was digested with Bsu36I/SphI and a 705 bp fragment containing the โ€˜3 region of the HSA encoding gene, nucleotide sequence encoding a GS linker, nucleotide sequence encoding human IL1-RA (N84Q) and the 5โ€™ region of a modified S.
  • cerevisiae ADH1 terminator (SEQ 1D3) was purified using standard techniques then ligated into Bsu36I/SphI-digested pDB4006 (containing HSA K573A expression cassette), pDB4010 (containing HSA D550N expression cassette), pDB4086 (containing HSA K500A expression cassette), pDB4110 (containing HSA K573P expression cassette) to generate pDB4287, pDB4286, pDB4285 and pDB4288, respectively (for an example, see FIG. 23 ).
  • pDB4285-pDB4288 were digested with NsiI/PvuI and the linearised DNA molecules were purified using standard techniques.
  • NsiI/PvuI-digested DNA samples were mixed with 100 ng Acc65I/BamHI-digested pDB3936 (9721 bp) (i.e. in vivo cloning) and used to transform S. cerevisiae (i.e. by in vivo cloning) using the Sigma Yeast Transformation kit described below.
  • the fusion polypeptides were analysed for their binding to FcRn using the SPR method described above and following results were obtained:
  • Example 8 it was shown that the K500A variant did not significantly bind shFcRn, in Example 10 it was shown that the K573P and K573A variants bind shFcRn stronger than HSA and in Example 11 it was shown that the D550N variant binds FcRn weaker than HSA.
  • fusion polypeptides in different configurations: C-terminal fusions with a small moiety (HSA-FLAG), C-terminal fusions with a larger polypeptide (HSA-IL1RA); N-terminal fusions with polypeptide (scFv-HSA); N- and C-terminal fusions (scFv-HSA-FLAG and scFv-HSA-scFv-FLAG).
  • recombinant albumin for conjugation analysis, commercially available recombinant albumin (RecombuminTM) was used as a control molecule.
  • RecombuminTM recombinant albumin
  • a final 200 mg/mL albumin K573P variant of the invention was purified from a fed batch fermentation by means described in Material and Methods. A two step purification was carried out;
  • the first step used a column (bed volume approximately 400 mL, bed height 11 cm) packed with AlbuPureTM matrix (ProMetic). This was equilibrated with 50 mM sodium acetate, pH 5.3 and loaded with neat culture supernatant, at approximately pH 5.5-6.5, to approximately 20 mg/mL matrix. The column was then washed with approximately 5 column volumes each of 50 mM sodium acetate, pH 5.3, 50 mM sodium phosphate, pH 6.0, 50 mM sodium phosphate, pH 7.0 and 50 mM ammonium acetate, pH 8.0, respectively. Bound protein was eluted using approximately two column volumes of 50 mM ammonium acetate, 10 mM octanoate, pH 7.0. The flow rate for the entire purification was 154 mL/min.
  • the eluate from the first step was diluted approximately two fold with water to give a conductivity of 2.5 โ‡ 0.5 mS/cm after adjustment to pH 5.5 โ‡ 0.3 with acetic acid.
  • This was loaded onto a DEAE-Sepharose Fast Flow (GE Healthcare) column (bed volume approximately 400 mL, bed height 11 cm), equilibrated with 80 mM sodium acetate, 5 mM octanoate, pH 5.5. Loading was approximately 30 mg protein/mL matrix.
  • the column was washed with approximately 5 column volumes of 80 mM sodium acetate, 5 mM octanoate, pH 5.5.
  • the eluate was concentrated and diafiltered against 145 mM NaCl, using a Pall Centramate Omega 10,000 Nominal MWCO membrane, to give a final protein concentration of approximately 200 mg/mL.
  • Both 200 mg/mL stock solutions of the rHA and K573P variant albumin were diluted down to 5 mg/mL, using phosphate buffer saline (PBS), pH adjusted to pH 6.5-6.7. This ensured a favourable pH environment for the maleimide reactive group of the EZ-Linkยฎ Maleimide Activated Horseradish Peroxidase (Thermo Scientific) to react with the free sulphydryl, to form a stable thioester bond.
  • 2 mg of the EZ-Linkยฎ Maleimide Activated Horseradish Peroxidase (HRP) was mixed with either 1 mL of the 5 mg/ML rHA or K573P variant albumin. This mixture ensured an approximate 2 fold molar excess of the albumin, or K573P variant albumin. This mixture was minimally incubated at 4ยฐ C., for 24 hours. The reaction mixtures were then checked for conjugation, using GP-HPLC.
  • the two same albumin samples used in Example 17, were also the start materials for this example. I.e. Approximately 200 mg/mL rHA or the K573P albumin variant.
  • Fluorescein-5-Maleimide was dissolved in dimethylformamide, to give a final concentration of 25 mg/mL. This was then further diluted into 18 mls of PBS, pH adjusted to approximately pH 6.5. To this solution either 1 ml of 200 mg/mL rHA or 1 mL of 200 mg/mL K573P variant was added. This gave an approximate 20 fold final molar excess of F5M. These samples were incubated and allowed to conjugate overnight at 4ยฐ C., in the dark, to allow the maleimide groups on the F5M to react with predominantly the free sulfhydryl, present in both albumin species.
  • FIGS. 30A and 30B shows the effect on shFcRn binding for the albumin variants. Substitutions N503D, D494N+E495Q+T496A E492T+N503D, E495Q+T496A within HSA had a negative impact on binding to shFcRn at pH5.5.
  • the competitive binding hierarchy was identical for the variants fusions of HSA-FLAG and, N+C-terminal scFv HSA-FLAG to the hierarchy of the individual HSA variants (unfused and fused) affinity data.
  • the IL1 Ra variants K573P, K573A, and the K500A were as predicted, however the D550N appears to inhibit more efficiently than the WT fusion.
  • HSA E492G+K573A HSA E492G+N503K+K573A
  • HSA E492G+N503H+K573A HSA E492G+K573P
  • HSA E492G+N503K+K573P HSA E492G+N503H+K573P.
  • SPR analysis was performed as described in Materials and Methods. Results ( FIG. 32 ) showed that all HSA variants bound more strongly to shFcRn compared to wild type HSA at pH 5.5. No binding was observed at pH 7.4.
  • HSA E492G+K573A HSA E492G+N503K+K573A, unlike HSA E492G+N503H+K573A, had marginally improved binding beyond that of HSA K573A.
  • the combination variants containing K573P did not show improved binding over the K573P single variant.

Abstract

The present invention relates to variants of a parent albumin having altered plasma half-life compared with the parent albumin. The present invention also relates to fusion polypeptides and conjugates comprising said variant albumin.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a division of U.S. application Ser. No. 14/863,719, filed Sep. 24, 2015, currently pending, which is a division of U.S. application Ser. No. 14/262,244 filed Apr. 25, 2014 (now abandoned), which is a division of U.S. application Ser. No. 13/504,326 filed Apr. 26, 2012 (now U.S. Pat. No. 8,748,380), which is a 35 U.S.C. 371 national application of PCT/EP2010/066572 filed Nov. 1, 2010, which designated the United States and was published in English and, which claims priority or the benefit under 35 U.S.C. 119 of European application nos. 10174162.7 and 09174698.2 filed Aug. 26, 2010 and Oct. 30, 2009, respectively, and U.S. provisional application Nos. 61/348,001 and 61/327,171 filed May 25, 2010 and Apr. 23, 2010, respectively. The contents of each of U.S. application Ser. No. 14/863,719, U.S. application Ser. No. 14/262,244, U.S. application Ser. No. 13/504,326, U.S. Pat. No. 8,748,380, International Application No. PCT/EP2010/066572, European application no. 10174162.7, European application no. 09174698.2, U.S. provisional application No. 61/348,001 and U.S. provisional application No. 61/327,171, are hereby expressly incorporated herein by reference in their entireties.
  • REFERENCE TO A SEQUENCE LISTING
  • This application contains a Sequence Listing in computer readable form, which is incorporated herein by reference.
  • BACKGROUND OF THE INVENTION Field of the Invention
  • The present invention relates to variants of albumin or fragments thereof or fusion polypeptides comprising variant albumin or fragments thereof having a change in half-life compared to the albumin, fragment thereof or fusion polypeptide comprising albumin or a fragment thereof.
  • Description of the Related Art
  • Albumin is a protein naturally found in the blood plasma of mammals where it is the most abundant protein. It has important roles in maintaining the desired osmotic pressure of the blood and also in transport of various substances in the blood stream.
  • Albumins have been characterized from many species including human, pig, mouse, rat, rabbit and goat and they share a high degree of sequence and structural homology.
  • Albumin binds in vivo to its receptor, the neonatal Fc receptor (FcRn) โ€œBrambellโ€ and this interaction is known to be important for the plasma half-life of albumin. FcRn is a membrane bound protein, expressed in many cell and tissue types. FcRn has been found to salvage albumin from intracellular degradation (Roopenian D. C. and Akilesh, S. (2007), Nat. Rev. Immunol 7, 715-725.). FcRn is a bifunctional molecule that contributes to maintaining a high level of IgGs and albumin in serum in mammals such as human beings.
  • Whilst the FcRn-immunoglobulin (IgG) interaction has been characterized in the prior art, the FcRn-albumin interaction is less well characterized. The major FcRn binding site is localized within DIII (381-585). Andersen et al (2010). Clinical Biochemistry 43, 367-372. Data indicates that IgG and albumin bind non-cooperatively to distinct sites on FcRn (Andersen et al. (2006), Eur. J. Immunol 36, 3044-3051; Chaudhury et al. (2006), Biochemistry 45, 4983-4990.).
  • It is known that mouse FcRn binds IgG from mice and humans whereas human FcRn appears to be more discriminating (Ober et al. (2001) Int. Immunol 13, 1551-1559). Andersen et al. (2010). Journal of Biological Chemistry 285(7):4826-36, describes the affinity of human and mouse FcRn for each mouse and human albumin (all possible combinations). No binding of albumin from either species was observed at physiological pH to either receptor. At acidic pH, a 100-fold difference in binding affinity was observed. In all cases, binding of albumin and IgG from either species to both receptors were additive.
  • Human serum albumin (HSA) has been well characterized as a polypeptide of 585 amino acids, the sequence of which can be found in Peters, T., Jr. (1996) All about Albumin: Biochemistry, Genetics and Medical, Applications pp 10, Academic Press, Inc., Orlando (ISBN 0-12-552110-3). It has a characteristic binding to its receptor FcRn, where it binds at pH 6.0 but not at pH 7.4.
  • The plasma half-life of HSA has been found to be approximately 19 days. A natural variant having lower plasma half-life has been identified (Peach, R. J. and Brennan, S. O., (1991) Biochim Biophys Acta. 1097:49-54) having the substitution D494N. This substitution generated an N-glycosylation site in this variant, which is not present in the wild-type albumin. It is not known whether the glycosylation or the amino acid change is responsible for the change in plasma half-life.
  • Albumin has a long plasma half-life and because of this property it has been suggested for use in drug delivery. Albumin has been conjugated to pharmaceutically beneficial compounds (WO 2000/69902A), and it was found that the conjugateโ€”maintained the long plasma half-life of albumin. The resulting plasma half-life of the conjugate was generally considerably longer than the plasma half-life of the beneficial therapeutic compound alone.
  • Further, albumin has been fused to therapeutically beneficial peptides (WO 2001/79271 A and WO 2003/59934 A) with the typical result that the fusion has the activity of the therapeutically beneficial peptide and a considerably longer plasma half-life than the plasma half-life of the therapeutically beneficial peptides alone.
  • Otagiri et al (2009), Biol. Pharm, Bull. 32(4), 527-534, discloses that 77 albumin variant are know, of these 25 are found in domain III. A natural variant lacking the last 175 amino acids at the carboxy termini has been shown to have reduced half-life (Andersen et al (2010), Clinical Biochemistry 43, 367-372). Iwao et al. (2007) studied the half-life of naturally accruing human albumin variants using a mouse model, and found that K541E and K560E had reduced half-life, E501K and E570K had increased half-life and K573E had almost no effect on half-life (Iwao, et. al. (2007) B.B.A. Proteins and Proteomics 1774, 1582-1590).
  • Galliano et al (1993) Biochim. Biophys. Acta 1225, 27-32 discloses a natural variant E505K. Minchiotti et al. (1990) discloses a natural variant K536E. Minchiotti et al (1987) Biochim. Biophys. Acta 916, 411-418 discloses a natural variant K574N. Takahashi et al (1987) Proc. Natl. Acad. Sci. USA 84, 4413-4417, discloses a natural variant D550G. Carlson et al (1992). Proc. Nat. Acad. Sci. USA 89, 8225-8229, discloses a natural variant D550A.
  • Albumin has the ability to bind a number of ligands and these become associated (associates) with albumin. This property has been utilized to extend the plasma half-life of drugs having the ability to noncovalently bind to albumin. This can also be achieved by binding a pharmaceutical beneficial compound, which has little or no albumin binding properties, to a moiety having albumin binding properties. See review article and reference therein, Kratz (2008). Journal of Controlled Release 132, 171-183.
  • Albumin is used in preparations of pharmaceutically beneficial compounds, in which such a preparation maybe for example, but not limited to, a nano particle or micro particle of albumin. In these examples the delivery of a pharmaceutically beneficial compound or mixture of compounds may benefit from alteration in the albumins affinity to its receptor where the beneficial compound has been shown to associate with albumin for the means of delivery.
  • It is not clear what determines the plasma half-life of the formed associates (for example but not limited to Levemirยฎ, Kurtzhals P et al. Biochem. J. 1995; 312:725-731) conjugates or fusion polypeptides but it appears to be a result of the combination of the albumin and the selected pharmaceutically beneficial compound/polypeptide. It would be desirable to be able to control the plasma half-life of given albumin conjugates, associates or albumin fusion polypeptides so that a longer or shorter plasma half-life can be achieved than given by the components of the association, conjugation or fusion, in order to be able to design a particular drug according to the particulars of the indication intended to be treated.
  • Albumin is known to accumulate and be catabolised in tumours, it has also been shown to accumulate in inflamed joints of rheumatoid arthritis sufferers. See review article and reference therein, Kratz (2008). Journal of Controlled Release 132, 171-183. It is envisaged that HSA variants with increased affinity for FcRn would be advantageous for the delivery of pharmaceutically beneficial compounds.
  • It may even be desirable to have variants of albumin that have little or no binding to FcRn in order to provide shorter half-lives or controlled serum pharmacokinetics as described by Kenanova et al (2009) J. Nucl. Med.; 50 (Supplement 2):1582).
  • SUMMARY OF THE INVENTION
  • The present invention provides variants of a parent albumin with improved properties compared to its parent. In particular the invention provides variants of a parent albumin having altered plasma half-life compare to its parent.
  • The present invention relates to isolated variants of albumin or fragments thereof, or fusion polypeptides comprising variant albumin or fragments thereof, of a parent albumin, comprising an alteration at one or more (several) positions corresponding to positions 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584 of the mature polypeptide of SEQ ID NO: 2, wherein the variant is not the variant consisting of SEQ ID NO: 2 with the substitution D494N, E501K, K541E, D550G,A, K573E or K574N.
  • The alteration at one or more position may independently be selected among substitutions, insertions and deletions, where substitution are preferred.
  • The present invention also relates to isolated polynucleotides encoding the variants; nucleic acid constructs, vectors, and host cells comprising the polynucleotides; and methods of producing the variants.
  • The present invention also relates to conjugates or associates comprising the variant albumin or fragment thereof according to the invention and a beneficial therapeutic moiety or to a fusion polypeptide comprising a variant albumin or fragment thereof of the invention and a fusion partner polypeptide.
  • The invention further relates to compositions comprising the variant albumin, fragment thereof, fusion polypeptide comprising variant albumin or fragment thereof or conjugates comprising the variant albumin or fragment thereof, according to the invention or associates comprising the variant albumin or fragment thereof, according to the invention. The compositions are preferably pharmaceutical compositions.
  • The invention further relates to a pharmaceutical composition comprising a variant albumin, fragment thereof, fusion polypeptide comprising variant albumin or fragment thereof or conjugates comprising the variant albumin or fragment thereof, or associates comprising the variant albumin or fragment thereof, wherein said variant albumin, fragment thereof, fusion polypeptide comprising variant albumin or fragment thereof or conjugates comprising the variant albumin or fragment or associates of variant albumin or fragment thereof has altered plasma half-life compared to the corresponding plasma half-life of the HSA or fragment thereof, fusion polypeptide comprising HSA or fragment thereof or conjugates or associates of HSA or, fragment thereof, comprising HSA or fragment thereof.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 shows a restriction map of the expression plasmid pDB4082.
  • FIG. 2 shows a restriction map of the expression plasmid pDB2305
  • FIG. 3 shows a restriction map of the expression plasmid pDB4005
  • FIG. 4 shows SPR sensorgrams 10 ฮผM albumin injected over shFcRn HSA (JTA)=fatty acid free HSA obtained from Sigma-Aldrich (A3782), HSA (Novozymes)=Commercial Recombinant human serum albumin (RECOMBUMIN).
  • FIG. 5 shows ELISA binding of shFcRn-GST to human serum albumin (HSA) variants (100-0.045 ฮผg/ml). Binding of WT, D494N, D494Q and D494A pH 6.0 and pH 7.4. Binding of WT, D494N, D494N/T496A and T496A at pH 6.0 and pH 7.4. Binding of WT, E495Q and E495A at pH 6.0 and pH 7.4.
  • FIG. 6 shows representative sensorgrams of binding of 0.2 ฮผM of HSA variants to immobilized shFcRn (หœ4600 RU). WT, D494N, D494Q, D494A, D494N/T496A and T496A.
  • FIG. 7 shows representative sensorgrams of binding of 1 ฮผM of HSA variants to immobilized shFcRn (หœ1400 RU). WT, D494N, D494Q, D494A, D494N/T496A and T496A.
  • FIG. 8 shows relative binding of the HSA variants compared to WT based on two independent SPR experiments as shown (A) FIG. 6 and (B) FIG. 7.
  • FIG. 9 shows ELISA: (A) binding of shFcRn to albumins from human, donkey, bovine, sheep, goat and rabbit at pH 6.0. (B) binding of shFcRn to albumin from guinea pig, hamster, rat and chicken at pH 6.0. (C) binding of shFcRn to albumin from human, donkey, bovine, sheep, goat and rabbit at pH 7.4. (D) binding of shFcRn to albumin from guinea pig, hamster, rat and chicken at pH 7.4. (E) relative binding of the different albumins. Relative binding of human albumin to shFcRn is defined as 1.0. The ELISA values represent the mean of duplicates.
  • FIG. 10 shows SPR: Binding of shFcRn-GST to albumin from several species at pH 6.0 and pH 7.4. Representative sensorgrams showing binding of 5.0 ฮผM of albumin from different species; (A) human, (B) donkey, (C) bovine, (D) goat, (E) sheep, (F) rabbit, (G) dog, (H) guinea pig, (I) hamster, (J) rat, (K) mouse and (L) chicken. The albumin variants were injected over immobilized GST-tagged shFcRn (หœ2100 RU). Injections were performed at 25ยฐ C. at a rate of 40 ฮผl/min.
  • FIG. 11 shows SPR sensorgrams of selected HSA mutants compared with wild-type HSA. 20 ฮผM of (A) WT and P499A (B) WT and K500A, (C) WT and K536A, (D) WT and P537A and (E) WT and K538A and (F) WT and K537A were injected over immobilized shFcRn at pH 6.0 (หœ1500 RU)
  • FIG. 12 shows SPR sensorgrams of HSA mutants compared with WT HSA. 10 ฮผM of (A) WT and K573A (B) WT and K573C, (C) WT and K573F, (D) WT and K573G and (E) WT and K573L and (F) WT and K573M, (G) WT and K573Q, (H) WT and K573R and (I) WT and K573T and (J) WT and K573V injected over immobilized shFcRn at pH 5.5 and pH7.4. Injections were performed at 25ยฐ C. at a flow rate of 80 ฮผl/min.
  • FIG. 13 shows SPR sensorgrams of HSA mutants compared with wild-type HSA. 10 ฮผM of (A) WT and K573D (B) WT and K573E, (C) WT and K573H, (D) WT and K5731 and (E) WT and K573N and (F) WT and K573P, (G) WT and K573S, (H) WT and K573* and (I) WT and K573W and (J) WT and K573Y injected over immobilized shFcRn at pH 5.5 and pH7.4. Injections were performed at 25ยฐ C. at a flow rate of 80 ฮผl/min.
  • FIG. 14 shows SPR sensorgrams of HSA mutants compared with wild-type HSA. 20 ฮผM of (A) WT and E492G+K538H+K541N+E542D (B) WT and E492T+N503K+K541A, (C) WT and E492P+N503K+K541G+E542P, (D) WT and E492H+E501P+N503H+E505D+T506S+T540S+K541E and (E) WT and A490D+E492T+V493L+E501P+N503D+A504E+E505K+T506F+K541D and (F) WT and E492G+V493P+K538H+K541N+E542D injected over immobilized shFcRn at pH 6.0. Injections were performed at 25ยฐ C. at a flow rate of 80 ฮผl/min.
  • FIG. 15 shows SPR sensorgrams of HSA mutants compared with wild-type HSA. Twenty ฮผM of (A) WT, (B) H440Q, (C) H464Q and (D) H535Q injected over immobilized shFcRn at pH 6.0. Injections were performed at 25ยฐ C. at a flow rate of 80 ฮผl/min.
  • FIG. 16 shows SPR sensorgrams of HSA mutant K500E compared with wild-type HSA. Ten ฮผM of HSA mutant K500E injected over immobilized shFcRn at pH 5.75. Injections were performed at 25ยฐ C. at a flow rate of 30 ฮผl/min.
  • FIG. 17 shows a restriction map of the expression plasmid pDB3017
  • FIG. 18 shows a restriction map of the expression plasmid pDB3021
  • FIG. 19 shows a restriction map of the expression plasmid pDB3056
  • FIG. 20 shows a restriction map of the expression plasmid pDB3165
  • FIG. 21 shows a restriction map of the expression plasmid pDB4172
  • FIG. 22 shows a restriction map of the expression plasmid pDB4267
  • FIG. 23 shows a restriction map of the expression plasmid pDB4285
  • FIG. 24 shows a GP-HPLC chromatogram of WT HSA and mutant K573P HRP conjugates for shFcRn analysis. Injections of 25 ฮผL were made onto a TSK G3000SWXL column (Tosoh Bioscience) as described in materials and methods.
  • FIG. 25 shows SDS PAGE separation followed by both visual (A) and ultraviolet (B) detection of the Fluorescein conjugated albumin. HSA::F5M (Lane 1), K573P::F5M (Lane 2) and rHA standard (Lane 3).
  • FIG. 26 shows shFcRn binding properties of HSA variants. 10 ฮผM of WT rHA and E492T(A), WT rHA and D494N/E495Q/T496A(B), WT rHA and N503D(C), WT rHA and N503K(D), WT rHA and E492T/N503D(E), WT rHA and E495Q/T496A(F), WT rHA and K538H(G), WT rHA and E492D(H) injected over immobilised shFcRn at pH5.5
  • FIG. 27 shows shFcRn binding properties of HSA variants. 10 ฮผM of WT rHA and K541A(I) and WT rHA and K541N(J) were injected over immobilised shFcRn at pH5.5.
  • FIG. 28 shows competitive binding of K573A and K573P measured by injecting shFcRn (100 nM) alone or pre-incubated with different amounts of HSA K573A and K573P over immobilized HSA (หœ2500 RU) at pH6.0
  • FIG. 29 shows competitive binding of HSA-FLAG variants measured by injecting shFcRn (100 nM) alone or together with different amounts of HSA-FLAG variants over immobilized HSA (หœ2500 RU) at pH6.0.
  • FIG. 30 shows competitive binding of HSA-IL1Ra variants measured by injecting shFcRn (100 nM) alone or together with different amounts of HSA-IL1Ra variants over immobilized HSA (หœ2500 RU) at pH6.0
  • FIG. 31 shows competitive binding of scFv-fused HSA variants measured by injecting shFcRn (หœ100 nM) alone or together with different amounts of (A) scFv-HSA-FLAG variants or (B) HSA-scFv-FLAG variants over immobilized HSA (หœ2500 RU) at pH6.0.
  • FIG. 32 shows binding of HSA, single, double and triple mutant variants to shFcRn. Samples of 10 ฮผM of each HSA variant were injected over immobilized shFcRn at pH 5.5 or pH 7.4.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention relates to isolated variants of albumin or fragments thereof, or fusion polypeptides comprising variant albumin or fragments thereof, of a parent albumin, comprising an alteration at one or more (several) positions corresponding to positions 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584 of the mature polypeptide of SEQ ID NO: 2, wherein the variant is not the variant consisting of SEQ ID NO: 2 with the substitution D494N, E501K, K541E, D550G,A, K573E or K574N.
  • The alteration at one or more position may independently be selected among substitutions, insertions and deletions, where substitution are preferred.
  • Definitions
  • Variant: The term โ€œvariantโ€ means a polypeptide derived from a parent albumin by one or more alteration(s), i.e., a substitution, insertion, and/or deletion, at one or more (several) positions. A substitution means a replacement of an amino acid occupying a position with a different amino acid; a deletion means removal of an amino acid occupying a position; and an insertion means adding 1 or more, preferably 1-3 amino acids immediately adjacent to an amino acid occupying a position.
  • Mutant: The term โ€œmutantโ€ means a polynucleotide encoding a variant.
  • Wild-Type Albumin: The term โ€œwild-typeโ€ (VVT) albumin means albumin having the same amino acid sequence as naturally found in an animal or in a human being.
  • Parent or Parent albumin The term โ€œparentโ€ or โ€œparent albuminโ€ means an albumin to which an alteration is made by the hand of man to produce the albumin variants of the present invention. The parent may be a naturally occurring (wild-type) polypeptide or an allele thereof, or even a variant thereof.
  • FcRn and shFcRn: The term โ€œFcRnโ€ means the human neonatal Fc receptor (FcRn). shFcRn is a soluble recombinant form of FcRn.
  • smFcRn: The term โ€œsmFcRnโ€ is a soluble recombinant form of the mouse neonatal Fc Receptor.
  • Isolated variant: The term โ€œisolated variantโ€ means a variant that is modified by the hand of man and separated completely or partially from at least one component with which it naturally occurs. In one aspect, the variant is at least 1% pure, e.g., at least 5% pure, at least 10% pure, at least 20% pure, at least 40% pure, at least 60% pure, at least 80% pure, and at least 90% pure, as determined by SDS-PAGE or GP-HPLC.
  • Substantially pure variant: The term โ€œsubstantially pure variantโ€ means a preparation that contains at most 10%, at most 8%, at most 6%, at most 5%, at most 4%, at most 3%, at most 2%, at most 1%, and at most 0.5% by weight of other polypeptide material with which it is natively or recombinantly associated. Preferably, the variant is at least 92% pure, e.g., at least 94% pure, at least 95% pure, at least 96% pure, at least 97% pure, at least 98% pure, at least 99%, at least 99.5% pure, and 100% pure by weight of the total polypeptide material present in the preparation. The variants of the present invention are preferably in a substantially pure form. This can be accomplished, for example, by preparing the variant by well known recombinant methods and by purification methods.
  • Mature polypeptide: The term โ€œmature polypeptideโ€ means a polypeptide in its final form following translation and any post-translational modifications, such as N-terminal processing, C-terminal truncation, glycosylation, phosphorylation, etc. In one aspect, the mature polypeptide is amino acids 1 to 585 of SEQ ID NO: 2, with the inclusion of any post-translational modifications.
  • Mature polypeptide coding sequence: The term โ€œmature polypeptide coding sequenceโ€ means a polynucleotide that encodes a mature albumin polypeptide. In one aspect, the mature polypeptide coding sequence is nucleotides 1 to 1758 of SEQ ID NO: 1.
  • Sequence Identity: The relatedness between two amino acid sequences or between two nucleotide sequences is described by the parameter โ€œsequence identityโ€.
  • For purposes of the present invention, the degree of sequence identity between two amino acid sequences is determined using the Needleman-Wunsch algorithm (Needleman and Wunsch, 1970, J. Mol. Biol. 48: 443-453) as implemented in the Needle program of the EMBOSS package (EMBOSS: The European Molecular Biology Open Software Suite, Rice et al., 2000, Trends Genet. 16: 276-277), preferably version 3.0.0 or later. The optional parameters used are gap open penalty of 10, gap extension penalty of 0.5, and the EBLOSUM62 (EMBOSS version of BLOSUM62) substitution matrix. The output of Needle labelled โ€œlongest identityโ€ (obtained using the -nobrief option) is used as the percent identity and is calculated as follows:

  • (Identical Residuesร—100)/(Length of Alignmentโˆ’Total Number of Gaps in Alignment)
  • For purposes of the present invention, the degree of sequence identity between two deoxyribonucleotide sequences is determined using the Needleman-Wunsch algorithm (Needleman and Wunsch, 1970, supra) as implemented in the Needle program of the EMBOSS package (EMBOSS: The European Molecular Biology Open Software Suite, Rice et al., 2000, supra), preferably version 3.0.0 or later. The optional parameters used are gap open penalty of 10, gap extension penalty of 0.5, and the EDNAFULL (EMBOSS version of NCBI NUC4.4) substitution matrix. The output of Needle labeled โ€œlongest identityโ€ (obtained using the -nobrief option) is used as the percent identity and is calculated as follows:

  • (Identical Deoxyribonucleotidesร—100)/(Length of Alignmentโˆ’Total Number of Gaps in Alignment)
  • Fragment: The term โ€œfragmentโ€ means a polypeptide having one or more (several) amino acids deleted from the amino and/or carboxyl terminus of an albumin and/or an internal region of albumin that has retained the ability to bind to FcRn. Fragments may consist of one uninterrupted sequence derived from HSA or it may comprise two or more sequences derived from HSA. The fragments according to the invention have a size of more than approximately 20 amino acid residues, preferably more than 30 amino acid residues, more preferred more than 40 amino acid residues, more preferred more than 50 amino acid residues, more preferred more than 75 amino acid residues, more preferred more than 100 amino acid residues, more preferred more than 200 amino acid residues, more preferred more than 300 amino acid residues, even more preferred more than 400 amino acid residues and most preferred more than 500 amino acid residues.
  • Allelic variant: The term โ€œallelic variantโ€ means any of two or more alternative forms of a gene occupying the same chromosomal locus. Allelic variation arises naturally through mutation, and may result in polymorphism within populations. Gene mutations can be silent (no change in the encoded polypeptide) or may encode polypeptides having altered amino acid sequences. An allelic variant of a polypeptide is a polypeptide encoded by an allelic variant of a gene.
  • Coding sequence: The term โ€œcoding sequenceโ€ means a polynucleotide, which directly specifies the amino acid sequence of its translated polypeptide product. The boundaries of the coding sequence are generally determined by an open reading frame, which usually begins with the ATG start codon or alternative start codons such as GTG and TTG and ends with a stop codon such as TAA, TAG, and TGA. The coding sequence may be a DNA, cDNA, synthetic, or recombinant polynucleotide.
  • cDNA: The term โ€œcDNAโ€ means a DNA molecule that can be prepared by reverse transcription from a mature, spliced, mRNA molecule obtained from a eukaryotic cell. cDNA lacks intron sequences that may be present in the corresponding genomic DNA. The initial, primary RNA transcript is a precursor to mRNA that is processed through a series of steps, including splicing, before appearing as mature spliced mRNA.
  • Nucleic acid construct: The term โ€œnucleic acid constructโ€ means a nucleic acid molecule, either single- or double-stranded, which is isolated from a naturally occurring gene or is modified to contain segments of nucleic acids in a manner that would not otherwise exist in nature or which is synthetic. The term nucleic acid construct is synonymous with the term โ€œexpression cassetteโ€ when the nucleic acid construct contains the control sequences required for expression of a coding sequence of the present invention.
  • Control sequences: The term โ€œcontrol sequencesโ€ means all components necessary for the expression of a polynucleotide encoding a variant of the present invention. Each control sequence may be native or foreign to the polynucleotide encoding the variant or native or foreign to each other. Such control sequences include, but are not limited to, a leader, polyadenylation sequence, propeptide sequence, promoter, signal peptide sequence, and transcription terminator. At a minimum, the control sequences include a promoter, and transcriptional and translational stop signals. The control sequences may be provided with linkers for the purpose of introducing specific restriction sites facilitating ligation of the control sequences within the coding region of the polynucleotide encoding a variant.
  • Operably linked: The term โ€œoperably linkedโ€ means a configuration in which a control sequence is placed at an appropriate position relative to the coding sequence of a polynucleotide such that the control sequence directs the expression of the coding sequence.
  • Expression: The term โ€œexpressionโ€ includes any step involved in the production of the variant including, but not limited to, transcription, post-transcriptional modification, translation, post-translational modification, and secretion.
  • Expression vector: The term โ€œexpression vectorโ€ means a linear or circular DNA molecule that comprises a polynucleotide encoding a variant and is operably linked to additional nucleotides that provide for its expression.
  • Host cell: The term โ€œhost cellโ€ means any cell type that is susceptible to transformation, transfection, transduction, and the like with a nucleic acid construct or expression vector comprising a polynucleotide of the present invention. The term โ€œhost cellโ€ encompasses any progeny of a parent cell that is not identical to the parent cell due to mutations that occur during replication.
  • Plasma half-life: Plasma half-life is ideally determined in vivo in suitable individuals. However, since it is time consuming and expensive and there inevitable are ethical concerns connected with doing experiments in animals or man it is desirable to use an in vitro assay for determining whether plasma half-life is extended or reduced. It is known that the binding of albumin to its receptor FcRn is important for plasma half-life and the correlation between receptor binding and plasma half-life is that a higher affinity of albumin to its receptor leads to longer plasma half-life. Thus for the present invention a higher affinity of albumin to FcRn is considered indicative of an increased plasma half-life and a lower affinity of albumin to its receptor is considered indicative of a reduced plasma half-life.
  • In this application and claims the binding of albumin to its receptor FcRn is described using the term affinity and the expressions โ€œstrongerโ€ or โ€œweakerโ€. Thus, it should be understood that a molecule having a higher affinity to FcRn than HSA is considered to bind stronger to FcRn than HSA and a molecule having a lower affinity to FcRn than HSA is considered to bind weaker to FcRn than HSA.
  • The terms โ€œlonger plasma half-lifeโ€ or โ€œshorter plasma half-lifeโ€ and similar expressions are understood to be in relationship to the corresponding parent albumin molecule. Thus, a longer plasma half-life with respect to a variant albumin of the invention means that the variant has longer plasma half-life than the corresponding albumin having the same sequences except for the alteration(s) in positions corresponding to 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584 in SEQ ID NO: 2.
  • Conventions for Designation of Variants
  • For purposes of the present invention, the mature polypeptide disclosed in SEQ ID NO: 2 is used to determine the corresponding amino acid residue in another albumin. The amino acid sequence of another albumin is aligned with the mature polypeptide disclosed in SEQ ID NO: 2, and based on the alignment, the amino acid position number corresponding to any amino acid residue in the mature polypeptide disclosed in SEQ ID NO: 2 is determined using the Needleman-Wunsch algorithm (Needleman and Wunsch, 1970, J. Mol. Biol. 48: 443-453) as implemented in the Needle program of the EMBOSS package (EMBOSS: The European Molecular Biology Open Software Suite, Rice et al., 2000, Trends Genet. 16: 276-277), preferably version 3.0.0 or later.
  • Identification of the corresponding amino acid residue in another albumin can be confirmed by an alignment of multiple polypeptide sequences using โ€œClustalWโ€ (Larkin et al., 2007, Bioinformatics 23: 2947-2948).
  • When the other polypeptide (or protein) has diverged from the mature polypeptide of SEQ ID NO: 2 such that traditional sequence-based comparison fails to detect their relationship (Lindahl and Elofsson, 2000, J. Mol. Biol. 295: 613-615), other pairwise sequence comparison algorithms can be used. Greater sensitivity in sequence-based searching can be attained using search programs that utilize probabilistic representations of polypeptide families (profiles) to search databases. For example, the PSI-BLAST program generates profiles through an iterative database search process and is capable of detecting remote homologs (Atschul et al., 1997, Nucleic Acids Res. 25: 3389-3402). Even greater sensitivity can be achieved if the family or superfamily for the polypeptide has one or more representatives in the protein structure databases. Programs such as GenTHREADER (Jones, 1999, J. Mol. Biol. 287: 797-815; McGuffin and Jones, 2003, Bioinformatics 19: 874-881) utilize information from a variety of sources (PSI-BLAST, secondary structure prediction, structural alignment profiles, and solvation potentials) as inputs to a neural network that predicts the structural fold for a query sequence. Similarly, the method of Gough et al., 2000, J. Mol. Biol. 313: 903-919, can be used to align a sequence of unknown structure within the superfamily models present in the SCOP database. These alignments can in turn be used to generate homology models for the polypeptide, and such models can be assessed for accuracy using a variety of tools developed for that purpose.
  • For proteins of known structure, several tools and resources are available for retrieving and generating structural alignments. For example the SCOP superfamilies of proteins have been structurally aligned, and those alignments are accessible and downloadable. Two or more protein structures can be aligned using a variety of algorithms such as the distance alignment matrix (Holm and Sander, 1998, Proteins 33: 88-96) or combinatorial extension (Shindyalov and Bourne, 1998, Protein Engineering 11: 739-747), and implementations of these algorithms can additionally be utilized to query structure databases with a structure of interest in order to discover possible structural homologs (e.g., Holm and Park, 2000, Bioinformatics 16: 566-567).
  • In describing the albumin variants of the present invention, the nomenclature described below is adapted for ease of reference. The accepted IUPAC single letter or three letter amino acid abbreviation is employed.
  • Substitutions. For an amino acid substitution, the following nomenclature is used: Original amino acid, position, substituted amino acid. Accordingly, for example the substitution of threonine with alanine at position 226 is designated as โ€œThr226Alaโ€ or โ€œT226Aโ€. Multiple mutations are separated by addition marks (โ€œ+โ€), e.g., โ€œGly205Arg+Ser411Pheโ€ or โ€œG205R+S411Fโ€, representing substitutions at positions 205 and 411 of glycine (G) with arginine (R) and serine (S) with phenylalanine (F), respectively. The Figures also use (โ€œ/โ€), e.g., โ€œE492T/N503Dโ€ this should be viewed as interchangeable with (โ€œ+โ€).
  • Deletions. For an amino acid deletion, the following nomenclature is used: Original amino acid, position*. Accordingly, the deletion of glycine at position 195 is designated as โ€œGly195*โ€ or โ€œG195*โ€. Multiple deletions are separated by addition marks (โ€œ+โ€), e.g., โ€œGly195*+Ser411*โ€ or โ€œG195*+S411*โ€.
  • Insertions. For an amino acid insertion, the following nomenclature is used: Original amino acid, position, original amino acid, inserted amino acid. Accordingly the insertion of lysine after glycine at position 195 is designated โ€œGly195GlyLysโ€ or โ€œG195GKโ€. An insertion of multiple amino acids is designated [Original amino acid, position, original amino acid, inserted amino acid #1, inserted amino acid #2; etc.]. For example, the insertion of lysine and alanine after glycine at position 195 is indicated as โ€œGly195GlyLysAlaโ€ or โ€œG195GKAโ€.
  • In such cases the inserted amino acid residue(s) are numbered by the addition of lower case letters to the position number of the amino acid residue preceding the inserted amino acid residue(s). In the above example, the sequence would thus be:
  • Parent: Variant:
    195 195 195a 195b
    G G-K-A
  • Multiple alterations. Variants comprising multiple alterations are separated by addition marks (โ€œ+โ€), e.g., โ€œArg170Tyr+Gly195Gluโ€ or โ€œR170Y+G195Eโ€ representing a substitution of tyrosine and glutamic acid for arginine and glycine at positions 170 and 195, respectively.
  • Different substitutions. Where different substitutions can be introduced at a position, the different substitutions are separated by a comma, e.g., โ€œArg170Tyr,Gluโ€ represents a substitution of arginine with tyrosine or glutamic acid at position 170. Thus, โ€œTyr167Gly,Ala+Arg170Gly,Alaโ€ designates the following variants: โ€œTyr167Gly+Arg170Glyโ€, โ€œTyr167Gly+Arg170Alaโ€, โ€œTyr167Ala+Arg170Glyโ€, and โ€œTyr167Ala+Arg170Alaโ€.
  • Parent Albumin
  • Albumins are proteins and constitute the most abundant protein in plasma in mammals and albumins from a long number of mammals have been characterized by biochemical methods and/or by sequence information. Several albumins, e.g., human serum albumin (HSA), have also been characterized crystallographically and the structure determined.
  • HSA is a preferred albumin according to the invention and is a protein consisting of 585 amino acid residues and has a molecular weight of 67 kDa. In its natural form it is not glycosylated. The amino acid sequence of HSA is shown in SEQ ID NO: 2. The skilled person will appreciate that natural alleles may exist having essentially the same properties as HSA but having one or more amino acid changes compared to SEQ ID NO: 2, and the inventors also contemplate the use of such natural alleles as parent albumin according to the invention.
  • Albumins have generally a long plasma half-life of approximately 20 days or longer, e.g., HSA has a plasma half-life of 19 days. It is known that the long plasma half-life of HSA is mediated via interaction with its receptor FcRn, however, an understanding or knowledge of the exact mechanism behind the long half-life of HSA is not essential for the present invention.
  • According to the invention the term โ€œalbuminโ€ means a protein having the same, or very similar three dimensional structure as HSA and having a long plasma half-life. As examples of albumin proteins according to the invention can be mentioned human serum albumin, primate serum albumin, (such as chimpanzee serum albumin, gorilla serum albumin), rodent serum albumin (such as hamster serum albumin, guinea pig serum albumin, mouse albumin and rat serum albumin), bovine serum albumin, equine serum albumin, donkey serum albumin, rabbit serum albumin, goat serum albumin, sheep serum albumin, dog serum albumin, chicken serum albumin and pig serum albumin. HSA as disclosed in SEQ ID NO: 2 or any naturally occurring allele thereof, is the preferred albumin according to the invention.
  • The parent albumin, a fragment thereof, or albumin part of a fusion polypeptide comprising albumin or a fragment thereof according to the invention has generally a sequence identity to the sequence of HSA shown in SEQ ID NO: 2 of at least 60%, preferably at least 70%, preferably at least 80%, preferably at least 85%, preferably at least 86%, preferably at least 87%, preferably at least 88%, preferably at least 89%, preferably at least 90%, preferably at least 91%, preferably at least 92%, preferably at least 93%, preferably at least 94%, preferably at least 95%, more preferred at least 96%, more preferred at least 97%, more preferred at least 98% and most preferred at least 99%.
  • The parent preferably comprises or consists of the amino acid sequence of SEQ ID NO: 2. In another aspect, the parent comprises or consists of the mature polypeptide of SEQ ID NO: 2.
  • In another embodiment, the parent is an allelic variant of the mature polypeptide of SEQ ID NO: 2.
  • In a second aspect, the parent is encoded by a polynucleotide that hybridizes under very low stringency conditions, low stringency conditions, medium stringency conditions, medium-high stringency conditions, high stringency conditions, or very high stringency conditions with (i) the mature polypeptide coding sequence of SEQ ID NO: 1, (ii) the mature polypeptide coding sequence of SEQ ID NO: 1, or (iii) the full-length complementary strand of (i) or (ii) (J. Sambrook, E. F. Fritsch, and T. Maniatis, 1989, Molecular Cloning, A Laboratory Manual, 2d edition, Cold Spring Harbor, N.Y.).
  • The polynucleotide of SEQ ID NO: 1 or a subsequence thereof, as well as the amino acid sequence of SEQ ID NO: 2 or a fragment thereof, may be used to design nucleic acid probes to identify and clone DNA encoding a parent from strains of different genera or species according to methods well known in the art. In particular, such probes can be used for hybridization with the genomic or cDNA of the genus or species of interest, following standard Southern blotting procedures, in order to identify and isolate the corresponding gene therein. Such probes can be considerably shorter than the entire sequence, but should be at least 14, e.g., at least 25, at least 35, or at least 70 nucleotides in length. Preferably, the nucleic acid probe is at least 100 nucleotides in length, e.g., at least 200 nucleotides, at least 300 nucleotides, at least 400 nucleotides, at least 500 nucleotides, at least 600 nucleotides, at least 700 nucleotides, at least 800 nucleotides, or at least 900 nucleotides in length. Both DNA and RNA probes can be used. The probes are typically labelled for detecting the corresponding gene (for example, with 32P, 3H, 35S, biotin, or avidin). Such probes are encompassed by the present invention.
  • A genomic DNA or cDNA library prepared from such other organisms may be screened for DNA that hybridizes with the probes described above and encodes a parent. Genomic or other DNA from such other organisms may be separated by agarose or polyacrylamide gel electrophoresis, or other separation techniques. DNA from the libraries or the separated DNA may be transferred to and immobilized on nitrocellulose or other suitable carrier material. In order to identify a clone or DNA that is homologous with SEQ ID NO: 1 or a subsequence thereof, the carrier material is used in a Southern blot.
  • For purposes of the present invention, hybridization indicates that the polynucleotide hybridizes to a labelled nucleotide probe corresponding to the polynucleotide shown in SEQ ID NO: 1, its complementary strand, or a subsequence thereof, under low to very high stringency conditions. Molecules to which the probe hybridizes can be detected using, for example, X-ray film or any other detection means known in the art.
  • In one aspect, the nucleic acid probe is the mature polypeptide coding sequence of SEQ ID NO: 1. In another aspect, the nucleic acid probe is nucleotides 1 to 1785 of SEQ ID NO: 1. In another aspect, the nucleic acid probe is a polynucleotide that encodes the polypeptide of SEQ ID NO: 2 or a fragment thereof. In another aspect, the nucleic acid probe is SEQ ID NO: 1.
  • For long probes of at least 100 nucleotides in length, very low to very high stringency conditions are defined as prehybridization and hybridization at 42ยฐ C. in 5ร—SSPE, 0.3% SDS, 200 micrograms/ml sheared and denatured salmon sperm DNA, and either 25% formamide for very low and low stringencies, 35% formamide for medium and medium-high stringencies, or 50% formamide for high and very high stringencies, following standard Southern blotting procedures for 12 to 24 hours optimally. The carrier material is finally washed three times each for 15 minutes using 2ร—SSC, 0.2% SDS at 45ยฐ C. (very low stringency), 50ยฐ C. (low stringency), 55ยฐ C. (medium stringency), 60ยฐ C. (medium-high stringency), 65ยฐ C. (high stringency), or 70ยฐ C. (very high stringency).
  • For short probes that are about 15 nucleotides to about 70 nucleotides in length, stringency conditions are defined as prehybridization and hybridization at about 5ยฐ C. to about 10ยฐ C. below the calculated Tm using the calculation according to Bolton and McCarthy (1962, Proc. Natl. Acad. Sci. USA 48: 1390) in 0.9 M NaCl, 0.09 M Tris-HCl pH 7.6, 6 mM EDTA, 0.5% NP-40, 1ร—Denhardt's solution, 1 mM sodium pyrophosphate, 1 mM sodium monobasic phosphate, 0.1 mM ATP, and 0.2 mg of yeast RNA per ml following standard Southern blotting procedures for 12 to 24 hours optimally. The carrier material is finally washed once in 6ร—SCC plus 0.1% SDS for 15 minutes and twice each for 15 minutes using 6ร—SSC at 5ยฐ C. to 10ยฐ C. below the calculated Tm.
  • In a third aspect, the parent is encoded by a polynucleotide with a sequence identity to the mature polypeptide coding sequence of SEQ ID NO: 1 of at least 60%, e.g., at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%, which encodes a polypeptide which is able to function as an albumin. In an embodiment, the parent is encoded by a polynucleotide comprising or consisting of SEQ ID NO: 1.
  • Preparation of Variants
  • In a further aspect the invention relates to a method for preparing a variant albumin, fragment thereof, or fusion polypeptide comprising variant albumin or a fragment thereof comprising the steps of:
      • a. Identifying one or more amino acid residue positions being important for the binding of albumin to FcRn, in an albumin or a fragment thereof or the albumin part of a fusion polypeptide comprising albumin or a fragment thereof;
      • b. Providing a nucleic acid encoding said albumin, the fragment thereof or the albumin part of a fusion polypeptide comprising albumin or the fragment thereof;
      • c. Modifying the nucleic acid provided in b., so that the one or more (several) amino acid residue located at the positions identified in a., are deleted or substituted or inserted with a different amino acid;
      • d. Expressing the modified nucleic acid in a suitable host cell; and
      • e. Recovering the variant albumin, the fragment thereof or the fusion polypeptide comprising variant albumin or the fragment thereof.
  • The identification of one or more amino acid residue positions being important for the binding of albumin to FcRn, in albumin, fragment thereof or the albumin part of a fusion polypeptide can be done in several ways including, but not limited to, random mutagenesis followed by analysis of the generated mutants and comparison with the non-mutated parent molecule, and identification based on structural considerations optionally followed by generation of variants having the identified alterations and comparison with the non-mutated patent molecule.
  • A preferred method for identification of one or more amino acid residue positions to be changed to in order to prepare a variant HSA having an altered binding to FcRn compared with natural HSA, comprises the following steps:
      • i) Identifying a non-human albumin having a different binding property to FcRn;
      • ii) Identifying the amino acid residues of the human serum albumin interacting with FcRn;
      • iii) Comparing the primary and/or the tertiary structure of the identified non-human albumin and human serum albumin with respect to the amino acid residues identified in step ii) and identifying the amino acid residues that differ between said non-human albumin and human serum albumin as being responsible for the observed binding difference; and
      • iv) Optionally preparing variants of HSA at the positions identified in step iii) and confirming that the prepared variants have altered binding to FcRn compared with HSA.
  • Step i) above may be done using the SPR assay described below. However, the skilled person will appreciate that other methods may be used to identify non-human albumins having different binding properties to FcRn than HSA, and that the method is not dependent on how the non-human albumin, having different binding properties to FcRn, has been identified.
  • In one preferred embodiment the identified non-human albumin has a stronger binding to FcRn than HSA. Examples of non-human albumins having stronger binding to FcRn than HSA include donkey serum albumin, rabbit serum albumin, dog serum albumin, hamster serum albumin, guinea pig serum albumin, mouse serum albumin and rat serum albumin. Step ii) may be accomplished by considering the structure of FcRn, HSA and the binding complex of these two. In the absence of an available structure of the binding complex it is possible to use a model where the HSA structure is docked into the structure of the FcRn structure and thereby identify amino acid residues of HSA interacting with FcRn.
  • In another preferred embodiment the identified non-human albumin has a weaker binding to FcRn than HSA. Examples of non-human albumins having weaker binding to FcRn than HSA include bovine serum albumin, goat serum albumin, sheep serum albumin and chicken serum albumin. Step ii) may be accomplished by considering the structure of FcRn, HSA and the binding complex of these two. In absence of an available structure of the binding complex it is possible to use a model where the HSA structure is docked into the structure of the FcRn structure and thereby identify residues of HSA interacting with FcRn.
  • In this invention and claims, an amino acid residues of HSA interacting with FcRn is considered any amino acid residues of HSA being located less than 10A from an amino acid in the FcRn or any amino acid residue that is involved in a hydrogen bond, a salt bridge or a polar or nonpolar interaction with an amino acid residue that is located less than 10A from an amino acid in the FcRn. Preferably the amino acid in HSA residues are located less than 10A from amino acids in the FcRn, more preferred less than 6A from amino acids in the FcRn and most preferred less than 3A from amino acids in the FcRn.
  • Step iii) and iv) can be done using techniques well known to the skilled person.
  • The present invention also relates to methods for obtaining a variant albumin or fragments thereof, or fusion polypeptides comprising the variant albumin or fragments thereof, or associates of variant albumin or fragment thereof comprising: (a) introducing into a parent albumin or fragments thereof, or fusion polypeptides comprising the parent albumin or fragments thereof an alteration at one or more (several) positions corresponding to positions 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584 of the mature polypeptide of SEQ ID NO: 2; and (b) recovering the variant albumin or fragments thereof, or fusion polypeptides comprising the variant albumin or fragments thereof.
  • The variants can be prepared by those skilled persons using any mutagenesis procedure known in the art, such as site-directed mutagenesis, synthetic gene construction, semi-synthetic gene construction, random mutagenesis, shuffling, etc.
  • Site-directed mutagenesis is a technique in which one or more (several) mutations are created at one or more defined sites in a polynucleotide encoding the parent.
  • Site-directed mutagenesis can be accomplished in vitro by PCR involving the use of oligonucleotide primers containing the desired mutation. Site-directed mutagenesis can also be performed in vitro by cassette mutagenesis involving the cleavage by a restriction enzyme at a site in the plasmid comprising a polynucleotide encoding the parent and subsequent ligation of an oligonucleotide containing the mutation in the polynucleotide. Usually the restriction enzyme that digests at the plasmid and the oligonucleotide is the same, permitting ligation of the plasmid and insert to one another. See, e.g., Scherer and Davis, 1979, Proc. Natl. Acad. Sci. USA 76: 4949-4955; and Barton et al., 1990, Nucleic Acids Res. 18: 7349-4966.
  • Site-directed mutagenesis can also be accomplished in vivo by methods known in the art. See, e.g., U.S. Patent Application Publication No. 2004/0171154; Storici et al., 2001, Nature Biotechnol. 19: 773-776; Kren et al., 1998, Nat. Med. 4: 285-290; and Calissano and Macino, 1996, Fungal Genet. Newslett. 43: 15-16.
  • Any site-directed mutagenesis procedure can be used in the present invention. There are many commercial kits available that can be used to prepare variants.
  • Synthetic gene construction entails in vitro synthesis of a designed polynucleotide molecule to encode a polypeptide of interest. Gene synthesis can be performed utilizing a number of techniques, such as the multiplex microchip-based technology described by Tian et al. (2004, Nature 432: 1050-1054) and similar technologies wherein oligionucleotides are synthesized and assembled upon photo-programmable microfluidic chips.
  • Single or multiple amino acid substitutions, deletions, and/or insertions can be made and tested using known methods of mutagenesis, recombination, and/or shuffling, followed by a relevant screening procedure, such as those disclosed by Reidhaar-Olson and Sauer, 1988, Science 241: 53-57; Bowie and Sauer, 1989, Proc. Natl. Acad. Sci. USA 86: 2152-2156; WO 95/17413; or WO 95/22625. Other methods that can be used include error-prone PCR, phage display (e.g., Lowman et al., 1991, Biochemistry 30: 10832-10837; U.S. Pat. No. 5,223,409; WO 92/06204) and region-directed mutagenesis (Derbyshire et al., 1986, Gene 46: 145; Ner et al., 1988, DNA 7: 127).
  • Mutagenesis/shuffling methods can be combined with high-throughput, automated screening methods to detect activity of cloned, mutagenized polypeptides expressed by host cells (Ness et al., 1999, Nature Biotechnology 17: 893-896). Mutagenized DNA molecules that encode active polypeptides can be recovered from the host cells and rapidly sequenced using standard methods in the art. These methods allow the rapid determination of the importance of individual amino acid residues in a polypeptide.
  • Semi-synthetic gene construction is accomplished by combining aspects of synthetic gene construction, and/or site-directed mutagenesis, and/or random mutagenesis, and/or shuffling. Semi-synthetic construction is typified by a process utilizing polynucleotide fragments that are synthesized, in combination with PCR techniques. Defined regions of genes may thus be synthesized de novo, while other regions may be amplified using site-specific mutagenic primers, while yet other regions may be subjected to error-prone PCR or non-error prone PCR amplification. Polynucleotide sub sequences may then be shuffled.
  • Variants
  • The present invention also provides variant albumins or fragments thereof, or fusion polypeptides comprising the variant albumin or fragments thereof, of a parent albumin, comprising an alteration at one or more (several) positions corresponding to positions 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584 in SEQ ID NO: 2, wherein each alteration is independently a substitution, insertion or deletion with the provision that the and the variant is not SEQ ID NO: 2 having the substitution D494N, E501K, K541E, D550G,A, K573E or K574N.
  • The variant albumin, a fragment thereof, or albumin part of a fusion polypeptide comprising variant albumin or a fragment thereof according to the invention has generally a sequence identity the sequence of HSA shown in SEQ ID NO: 2 of at least 60%, preferably at least 70%, preferably at least 80%, preferably at least 85%, preferably at least 90%, more preferred at least 95%, more preferred at least 96%, more preferred at least 97%, more preferred at least 98% and most preferred at least 99%.
  • In one aspect, the number of alterations in the variants of the present invention is 1-20, e.g., 1-10 and 1-5, such as 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 alterations.
  • The variant albumin, a fragment thereof or fusion polypeptide comprising the variant albumin or fragment thereof has altered plasma half-life compared with the corresponding parent albumin, fragment thereof, or fusion polypeptide comprising the variant albumin or fragment thereof.
  • In a particular preferred embodiment the parent albumin is HSA and the variant albumin, a fragment thereof or fusion polypeptide comprising the variant albumin or fragment thereof has altered plasma half-life compared with the HSA, the corresponding fragment or fusion polypeptide comprising HSA or fragment thereof.
  • The correlation between binding of albumin to its receptor and plasma half-life has been realized by the present inventors based on the natural occurring allele of HSA D494N. The inventors have analyzed this allele and found that it has a lower affinity to its receptor FcRn.
  • Further, it has been disclosed that a transgenic mouse having the natural mouse FcRn replaced with human FcRn has a higher serum albumin level than normal mouse; see (J Exp Med. (2003) 197(3):315-22). The inventors have discovered that human FcRn has a higher affinity to mouse serum albumin than mouse FcRn has to mouse serum albumin and, therefore, the observed increase in serum albumin in the transgenic mice corresponds with a higher affinity between serum albumin and its receptor, confirming the correlation between albumin binding to FcRn and plasma half-life. In addition, variants of albumin that have little or no binding to FcRn have been shown to have reduced half-life in a mouse model, Kenanova et al (2009) J. Nucl. Med.; 50 (Supplement 2): 1582).
  • One way to determine whether the affinity of a variant albumin to FcRn is higher or lower than the parent albumin is to use the Surface Plasmon Resonance assay (SPR) as described below. The skilled person will understand that other methods might be useful to determine whether the affinity of a variant albumin to FcRn is higher or lower than the affinity of the parent albumin to FcRn, e.g., determination and comparison of the binding constants KD. Thus, according to the invention variant albumins having a KD that is lower than the KD for natural HSA is considered to have a higher plasma half-life than HSA and variant albumins having a KD that is higher than the KD for natural HSA is considered to have a lower plasma half-life than HSA.
  • The variants of albumin or fragments thereof or fusion polypeptides comprising albumin or fragments thereof comprise one or more alterations, such as substitutions, deletions or insertions at one or more (several) positions corresponding to the positions in HSA selected from the group consisting of 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584. The substitution may be any substitution where the amino acid in the natural albumin sequence is substituted with a different amino acid selected among the remaining 19 natural occurring amino acids.
  • In one aspect, a variant comprises an alteration at one or more (several) positions corresponding to positions 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584 in SEQ ID NO: 2. In another aspect, a variant comprises an alteration at two positions corresponding to any of 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584 in SEQ ID NO: 2. In another aspect, a variant comprises an alteration at three positions corresponding to any of positions 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584 in SEQ ID NO: 2. In another aspect, a variant comprises an alteration at each position corresponding to positions 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584 in SEQ ID NO: 2.
  • In another aspect, the variant comprises the substitution Q417A,H of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution H440Q of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution H464Q of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution A490D of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution E492G, T,P,H of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution V493P,L of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution D494N,Q,A,E,P of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution E495Q,A of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution T496A of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution P499A of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution K500E,G,D,A,S,C,P,H,F,N,W,T,M,Y,V,Q,L,I,R of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution E501A,P,Q of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution N503K,D,H of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution A504E of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution E505K, D of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution T506F, S of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution H510Q of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution H535Q of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution K536A of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution P537A of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution K538A,H of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution T540S of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution K541A,D,G,N,E of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution E542P,D of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution D550N of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution K573Y,W,P,H,F,V,I,T,N,S,G,M,C,A,E,Q,R,L,D of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution K574N of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution Q580K of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution L575F of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution A577T,E of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution A578R,S of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution S579C,T of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution Q580K of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution A581D of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution A582T of the mature polypeptide of SEQ ID NO: 2. In another aspect, the variant comprises the substitution G584A of the mature polypeptide of SEQ ID NO: 2.
  • In one aspect, the variant comprises an alteration at a position corresponding to position 417. In another aspect, the amino acid at a position corresponding to position 417 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Ala or His. In another aspect, the variant comprises the substitution Q417A, H of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 440. In another aspect, the amino acid at a position corresponding to position 440 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Ala. In another aspect, the variant comprises the substitution H440Q of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 464. In another aspect, the amino acid at a position corresponding to position 464 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Ala. In another aspect, the variant comprises the substitution H464Q of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 490 In another aspect, the amino acid at a position corresponding to position 490 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val. In another aspect, the variant comprises the substitution A490G of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 492. In another aspect, the amino acid at a position corresponding to position 492 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Gly. In another aspect, the variant comprises the substitution E492G of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 493. In another aspect, the amino acid at a position corresponding to position 493 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Pro. In another aspect, the variant comprises the substitution V493P of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 494. In another aspect, the amino acid at a position corresponding to position 494 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Asn, Gin or Ala. In another aspect, the variant comprises the substitution D494N,Q,A of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 495. In another aspect, the amino acid at a position corresponding to position 495 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Gin or Ala. In another aspect, the variant comprises the substitution E495Q or A of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 496. In another aspect, the amino acid at a position corresponding to position 496 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Ala. In another aspect, the variant comprises the substitution T496A of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 499. In another aspect, the amino acid at a position corresponding to position 499 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Ala. In another aspect, the variant comprises the substitution P499A of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 500. In another aspect, the amino acid at a position corresponding to position 500 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Ala. In another aspect, the variant comprises the substitution K500E,G,D,A,S,C,P,H,F,N,W,T,M,Y,V,Q,L,I,R of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 501. In another aspect, the amino acid at a position corresponding to position 501 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Ala or Gln to reduce affinity and Pro to increase affinity. In another aspect, the variant comprises the substitution E501A, Q, P of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 503. In another aspect, the amino acid at a position corresponding to position 503 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Asp or Lys or His. In another aspect, the variant comprises the substitution N503D, K, H of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 504. In another aspect, the amino acid at a position corresponding to position 504 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val. In another aspect, the variant comprises the substitution A504 of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 505. In another aspect, the amino acid at a position corresponding to position 505 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val. In another aspect, the variant comprises the substitution E505D of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 506. In another aspect, the amino acid at a position corresponding to position 506 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val. In another aspect, the variant comprises the substitution T506S,F of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 510. In another aspect, the amino acid at a position corresponding to position 510 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Gln. In another aspect, the variant comprises the substitution H510Q of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 535. In another aspect, the amino acid at a position corresponding to position 535 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Gln. In another aspect, the variant comprises the substitution H535Q of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 536. In another aspect, the amino acid at a position corresponding to position 536 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Ala. In another aspect, the variant comprises the substitution K536A of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 537. In another aspect, the amino acid at a position corresponding to position 537 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Ala. In another aspect, the variant comprises the substitution P537A of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 538. In another aspect, the amino acid at a position corresponding to position 538 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Ala. In another aspect, the variant comprises the substitution K538H, A of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 540. In another aspect, the amino acid at a position corresponding to position 540 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val. In another aspect, the variant comprises the substitution T540S of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 541. In another aspect, the amino acid at a position corresponding to position 541 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Gly, Asp or Ala. In another aspect, the variant comprises the substitution K541G, D A, N of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 542. In another aspect, the amino acid at a position corresponding to position 542 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Asp or Pro. In another aspect, the variant comprises the substitution E542D, P of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 550. In another aspect, the amino acid at a position corresponding to position 550 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Asn to reduce affinity, preferably with Glu to increase affinity.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 573. In another aspect, the amino acid at a position corresponding to position 573 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Tyr, Trp, Pro, His. Phe, Val, Ile, Thr, Asn, Ser, Gly, Met, Cys, Ala, Glu, Gln, Arg, Leu, Asp. In another aspect, the variant comprises the substitution K573Y,W,P,H,F,V,I,T,N,S,G,M,C,A,E,Q,R,L,D of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 574. In another aspect, the amino acid at a position corresponding to position 574 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Asn. In another aspect, the variant comprises the substitution K574N of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 575. In another aspect, the amino acid at a position corresponding to position 575 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Phe. In another aspect, the variant comprises the substitution L575F of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 577. In another aspect, the amino acid at a position corresponding to position 577 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Thr or Glu. In another aspect, the variant comprises the substitution A577TE of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 578. In another aspect, the amino acid at a position corresponding to position 578 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Arg or Ser. In another aspect, the variant comprises the substitution A578R,S of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 579. In another aspect, the amino acid at a position corresponding to position 579 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Cys or Thr. In another aspect, the variant comprises the substitution S579C,T of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 580. In another aspect, the amino acid at a position corresponding to position 580 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Lys. In another aspect, the variant comprises the substitution Q580K of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 581. In another aspect, the amino acid at a position corresponding to position 581 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Asp. In another aspect, the variant comprises the substitution A581D of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 582. In another aspect, the amino acid at a position corresponding to position 582 is substituted with Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Thr. In another aspect, the variant comprises the substitution A582T of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at a position corresponding to position 584. In another aspect, the amino acid at a position corresponding to position 584 is substituted with Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or Val, preferably with Ala. In another aspect, the variant comprises the substitution G584A of the mature polypeptide of SEQ ID NO: 2.
  • In another aspect, the variant comprises an alteration at positions corresponding to positions 494 and 496 in SEQ ID NO: 2, such as those described above.
  • In another aspect, the variant comprises alterations at positions corresponding to positions 492 and 493 in SEQ ID NO: 2, such as those described above.
  • In another aspect, the variant comprises alterations at positions corresponding to positions 494 and 417 in SEQ ID NO: 2, such as those described above.
  • In another aspect, the variant comprises alterations at positions corresponding to positions 492 and 503 in SEQ ID NO: 2, such as those described above.
  • In another aspect, the variant comprises alterations at positions corresponding to positions 492 and 573 in SEQ ID NO: 2, such as those described above.
  • In another aspect, the variant comprises alterations at positions corresponding to positions 492, 503, and 573 in SEQ ID NO: 2, such as those described above.
  • In one embodiment the variant albumin or fragments thereof, or fusion polypeptides comprising the variant albumin or fragments thereof according to the invention contains one substitution at a position corresponding to a position in HSA selected from the group consisting of 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584 in SEQ ID NO: 2 provided that the variant albumin is not the variant consisting of SEQ ID NO: 2 with the substitution D494N, E501K, K541E, D550G,A, K573E or K574N. The variant albumin, fragment thereof or fusion polypeptides comprising variant albumin or a fragment thereof according to the invention may comprise additional substitutions, insertions or deletions at one or more (several) positions corresponding to other positions in HSA.
  • In another embodiment the variant albumin or fragments thereof, or fusion polypeptides comprising variant albumin or fragments thereof according to the invention contains two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen fifteen, sixteen, seventeen, eighteen, nineteen twenty or even more substitutions at positions corresponding to positions in HSA selected from the group consisting of 417, 440, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 575, 577, 578, 579, 580, 581, 582 and 584 of SEQ ID NO: 2. The variant albumin or fragments thereof, or fusion polypeptides comprising variant albumin or fragments thereof according to the invention may comprise additional substitutions, insertions or deletions at positions corresponding to other positions in HSA.
  • In a further embodiment the variants of albumin or fragments thereof, or fusion polypeptides comprising variant albumin or a fragment thereof according to the invention have a plasma half-life that is longer than the plasma half-life of the parent albumin fragment thereof or fusion polypeptide comprising the parent albumin or a fragment thereof. Examples according to this embodiment include variants of albumin or fragments thereof, or fusion polypeptides comprising variant albumin or a fragment thereof comprising a substitution in the position corresponding to 492, 503, 542, 550, 573, 574, 580, 581, 582 or 584 in SEQ ID NO: 2. Preferred substitutions according to this embodiment of the invention include the substitution of the amino acid residue in the position corresponding to 492 in SEQ ID NO: 2 with a G residue, substitution of the amino acid residue in the position corresponding to 503 in SEQ ID NO: 2 with a H or a K residue, substitution of the amino acid residue in the position corresponding to 550 in SEQ ID NO: 2 with an E residue, the substitution of the amino acid residue in a position corresponding to 573 in SEQ ID NO: 2 with an Y,W,P,H,F,V,I,T,N,S,G,M,C,A,E,Q,R,L or a D, the substitution of the amino acid residue in a position corresponding to 574 in SEQ ID NO: 2 with an N residue, or the substitution of the amino acid residue in the position corresponding to 580 in SEQ ID NO: 2 with an K residue. Other preferred variants have a substitution in the position corresponding to 492 in SEQ ID NO: 2 with a G residue and a substitution in the position corresponding to 573 in SEQ ID NO: 2 with an A or a P residue. Other preferred variant has a number of substitutions corresponding to position 492 in SEQ ID NO: 2 with an H residue in position 503 in SEQ ID NO: 2.
  • Other preferred variants have a substitution in the position corresponding to 492 in SEQ ID NO: 2 with a G residue and a substitution in the position corresponding to position 503 in SEQ ID NO: 2 corresponding to a H or a K and a substitution in position 573 in SEQ ID NO: 2 with an A or a P residue.
  • In a further embodiment the variants of albumin or fragments thereof, or fusion polypeptides comprising variant albumin or fragments thereof according to the invention have a plasma half-life that is shorter than the plasma half-life of the parent albumin fragment thereof or fusion polypeptide comprising the parent albumin or a fragment thereof. Examples according to this embodiment include variants of albumin or fragments thereof, or fusion polypeptides comprising variant albumin or a fragment thereof comprising a substitution in the position corresponding to 417, 440, 494, 495, 496, 499, 500, 501, 536, 537, 538, 541, 494+496 or 492+493 in SEQ ID NO: 2. Preferred substitutions include the substitutions corresponding to Q417A, H440Q, D494E+Q417H, D494N,Q,A, E495Q,A, T496A, D494N+T496A or, P499A, K500A, E501A, E501Q, K536A, P537A, K538A, K541G, K541A K541D or D550N in SEQ ID NO: 2.
  • In another embodiment of the invention the variants of albumin or fragments thereof, or fusion polypeptides comprising variant albumin or a fragment thereof according to the invention have lost their ability to bind FcRn. In this connection variants of albumin or fragments thereof, or fusion polypeptides comprising variant albumin or fragments thereof is considered to have lost the ability to bind FcRn if the measured resonance units for the variant in the SPR assay described below is less than 10% of the measured resonance units for the corresponding parent albumin or fragment thereof.
  • Examples according to this embodiment include variants of albumin or fragments thereof, or fusion polypeptides comprising variant albumin or fragments thereof comprising a substitution at a position corresponding to 464, 500, 510 or 535 in SEQ ID NO: 2. Preferred substitutions include the substitutions corresponding to H464Q, K500A,P,C,S,A,D.G H510Q or H535Q in SEQ ID NO: 2. In addition to the one or more substitutions at one or more positions corresponding to positions 417, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574, 580 581, 582 and 584 in SEQ ID NO: 2 the variant albumin or fragments thereof, or fusion polypeptides comprising variant albumin or fragments thereof according to the invention may contain additional substitutions, deletions or insertions in other positions of the molecules. Such additional substitutions, deletions or insertions may be useful in order to alter other properties of the molecules such as but not limited to altered glycosylation; introduction of reactive groups of the surface such a thiol groups, removing/generating a carbamoylation site; etc.
  • Residues that might be altered in order to provide reactive residues on the surface and which advantageously could be applied to the present invention has been disclosed in the unpublished patent application WO 2010/092135 (Included by reference). Particular preferred residues include the positions corresponding to positions in SEQ ID NO: 2.
  • As examples of alterations that can be made in SEQ ID NO: 2 or in corresponding positions in other albumins in order to provide a reactive thiol group on the surface includes alterations corresponding to following alterations in SEQ ID NO: 2: L585C, D1C, A2C, D562C, A364C, A504C, E505C, T79C, E86C, D129C, D549C, A581C, D121C, E82C, S270C, A578C, L595LC, D1DC, A2AC, D562DC, A364AC, A504AC, E505EC, T79TC, E86EC, D129DC, D549DC, A581AC, A581AC, D121DC, E82EC, S270SC, A579AC, C360*, C316*, C75*, C168*, C558*, C361*, C91*, C124*, C169* and C567*. Alternatively a cysteine residue may be added to the N or C terminal of albumin.
  • Polynucleotides
  • The present invention also relates to isolated polynucleotides that encode any of the variants of the present invention.
  • Nucleic Acid Constructs
  • The present invention also relates to nucleic acid constructs comprising a polynucleotide encoding a variant of the present invention operably linked to one or more (several) control sequences that direct the expression of the coding sequence in a suitable host cell under conditions compatible with the control sequences.
  • A polynucleotide may be manipulated in a variety of ways to provide for expression of a variant. Manipulation of the polynucleotide prior to its insertion into a vector may be desirable or necessary depending on the expression vector. The techniques for modifying polynucleotides utilizing recombinant DNA methods are well known in the art.
  • The control sequence may be a promoter sequence, which is recognized by a host cell for expression of the polynucleotide. The promoter sequence contains transcriptional control sequences that mediate the expression of the variant. The promoter may be any nucleic acid sequence that shows transcriptional activity in the host cell including mutant, truncated, and hybrid promoters, and may be obtained from genes encoding extracellular or intracellular polypeptides either homologous or heterologous to the host cell.
  • In a yeast host, useful promoters are obtained from the genes for Saccharomyces cerevisiae enolase (ENO-1), Saccharomyces cerevisiae protease A (PRA1), Saccharomyces cerevisiae protease B (PRB1), Saccharomyces cerevisiae translation elongation factor (TEF1), Saccharomyces cerevisiae translation elongation factor (TEF2), Saccharomyces cerevisiae galactokinase (GAL1), Saccharomyces cerevisiae alcohol dehydrogenase/glyceraldehyde-3-phosphate dehydrogenase (ADH1, ADH2/GAP), Saccharomyces cerevisiae triose phosphate isomerase (TPI), Saccharomyces cerevisiae metallothionein (CUP1), and Saccharomyces cerevisiae 3-phosphoglycerate kinase. Other useful promoters for yeast host cells are described by Romanos et al., 1992, Yeast 8: 423-488.
  • The control sequence may also be a suitable transcription terminator sequence, which is recognized by a host cell to terminate transcription. The terminator sequence is operably linked to the 3โ€ฒ-terminus of the polynucleotide encoding the variant. Any terminator that is functional in the host cell may be used.
  • Preferred terminators for yeast host cells are obtained from the genes for Saccharomyces cerevisiae enolase, Saccharomyces cerevisiae cytochrome C (CYC1), Saccharomyces cerevisiae alcohol dehydrogenase (ADH1) and Saccharomyces cerevisiae glyceraldehyde-3-phosphate dehydrogenase. Other useful terminators for yeast host cells are described by Romanos et al., 1992, supra.
  • The control sequence may also be a suitable leader sequence, a nontranslated region of an mRNA that is important for translation by the host cell. The leader sequence is operably linked to the 5โ€ฒ-terminus of the polynucleotide encoding the variant. Any leader sequence that is functional in the host cell may be used.
  • Suitable leaders for yeast host cells are obtained from the genes for Saccharomyces cerevisiae enolase (ENO-1), Saccharomyces cerevisiae 3-phosphoglycerate kinase, Saccharomyces cerevisiae alpha-factor, and Saccharomyces cerevisiae alcohol dehydrogenase/glyceraldehyde-3-phosphate dehydrogenase (ADH2/GAP).
  • The control sequence may also be a polyadenylation sequence, a sequence operably linked to the 3โ€ฒ-terminus of the variant-encoding sequence and, when transcribed, is recognized by the host cell as a signal to add polyadenosine residues to transcribed mRNA. Any polyadenylation sequence that is functional in the host cell may be used.
  • Useful polyadenylation sequences for yeast host cells are described by Guo and Sherman, 1995, Mol. Cellular Biol. 15: 5983-5990.
  • The control sequence may also be a signal peptide coding region that encodes a signal peptide linked to the N-terminus of a variant and directs the variant into the cell's secretory pathway. The 5โ€ฒ-end of the coding sequence of the polynucleotide may inherently contain a signal peptide coding region naturally linked in translation reading frame with the segment of the coding region that encodes the variant. Alternatively, the 5โ€ฒ-end of the coding sequence may contain a signal peptide coding region that is foreign to the coding sequence. The foreign signal peptide coding region may be required where the coding sequence does not naturally contain a signal peptide coding region. Alternatively, the foreign signal peptide coding region may simply replace the natural signal peptide coding region in order to enhance secretion of the variant. However, any signal peptide coding region that directs the expressed variant into the secretory pathway of a host cell may be used.
  • Useful signal peptides for yeast host cells are obtained from the genes for Saccharomyces cerevisiae alpha-factor and Saccharomyces cerevisiae invertase. Other useful signal peptide coding sequences are described by Romanos et al., 1992, supra.
  • Where both signal peptide and propeptide regions are present at the N-terminus of a variant, the propeptide region is positioned next to the N-terminus of the variant and the signal peptide region is positioned next to the N-terminus of the propeptide region.
  • Methods of Production
  • The variants of the present invention can be prepared using techniques well known to the skilled person. One convenient way is by cloning nucleic acid encoding the parent albumin or a fragment thereof or fusion polypeptide comprising albumin or a fragment thereof, modifying said nucleic acid to introduce the desired substitution(s) at one or more (several) positions corresponding to positions 417, 464, 490, 492, 493, 494, 495, 496, 499, 500, 501, 503, 504, 505, 506, 510, 535, 536, 537, 538, 540, 541, 542, 550, 573, 574 and 580 in SEQ ID NO: 2, where the variant is not the variant consisting of SEQ ID NO:2 with the substitution D494N, E501K, K541E, D550G,A, K573E or K574N., preparing a suitable genetic construct where the modified nucleic acid is placed in operative connection with suitable regulatory genetic elements, such as promoter, terminator, activation sites, ribosome binding sites etc., introducing the genetic construct into a suitable host organism, culturing the transformed host organism under conditions leading to expression of the variant and recovering the variant. All these techniques are known in the art and it is within the skills of the average practitioner to design a suitable method for preparing a particular variant according to the invention.
  • The variant polypeptide of the invention may also be connected to a signal sequence in order to have the variant polypeptide secreted into the growth medium during culturing of the transformed host organism. It is generally advantageous to have the variant polypeptide secreted into the growth medium in order to ease recovery and purification.
  • Techniques for preparing variant polypeptides have also been disclosed in WO 2009019314 (included by reference) and these techniques may also be applied to the present invention.
  • Albumins have been successfully expressed as recombinant proteins in a range of hosts including fungi (including but not limited to Aspergillus (WO06066595), Kluyveromyces (Fleer 1991, Bio/technology 9, 968-975), Pichia (Kobayashi 1998 Therapeutic Apheresis 2, 257-262) and Saccharomyces (Sleep 1990, Bio/technology 8, 42-46)), bacteria (Pandjaitab 2000, J. Allergy Clin. Immunol. 105, 279-285)), animals (Barash 1993, Transgenic Research 2, 266-276) and plants (including but not limited to potato and tobacco (Sijmons 1990, Bio/technology 8, 217 and Farran 2002, Transgenic Research 11, 337-346). The variant polypeptide of the invention is preferably produced recombinantly in a suitable host cell. In principle any host cell capable of producing a polypeptide in suitable amounts may be used and it is within the skills of the average practitioner to select a suitable host cell according to the invention. A preferred host organism is yeast, preferably selected among Saccharomycacae, more preferred Saccharomyces cerevisiae.
  • The variant polypeptides of the invention may be recovered and purified from the growth medium using a combination of known separation techniques such as filtration, centrifugation, chromatography, and affinity separation techniques etc. It is within the skills of the average practitioner to purify the variants of the invention using a particular combination of such known separation steps. As an example of purification techniques that may be applied to the variants of the present invention can be mentioned the teaching of WO0044772.
  • The variant polypeptides of the invention may be used for delivering a therapeutically beneficial compound to an animal or a human individual in need thereof. Such therapeutically beneficial compounds include, but are not limited, to labels and readily detectable compounds for use in diagnostics, such as various imaging techniques; pharmaceutical active compounds such as drugs, or specifically binding moieties such as antibodies. The variants of the invention may even be connected to two or more different therapeutically beneficial compounds, e.g., an antibody and a drug, which gives the combined molecule the ability to bind specifically to a desired target and thereby provide a high concentration of the connected drug at that particular target.
  • Fusion Polypeptides
  • The variants of albumin or fragments thereof according to the invention may also be fused with a non-albumin polypeptide fusion partner. The fusion partner may in principle be any polypeptide but generally it is preferred that the fusion partner is a polypeptide having therapeutic or diagnostic properties. Fusion polypeptides comprising albumin or fragments thereof are known in the art. It has been found that such fusion polypeptide comprising albumin or a fragment thereof and a fusion partner polypeptide have a longer plasma half-life compared to the unfused fusion partner polypeptide. According to the invention it is possible to alter the plasma half-life of the fusion polypeptides according to the invention compared to the corresponding fusion polypeptides of the prior art.
  • One or more therapeutic polypeptides may be fused to the N-terminus, the C-terminus of albumin, inserted into a loop in the albumin structure or any combination thereof. It may or it may not comprise linker sequences separating the various components of the fusion polypeptide.
  • Teachings relating to fusions of albumin or a fragment thereof are known in the art and the skilled person will appreciate that such teachings can also be applied to the present invention. WO 2001/79271 A and WO 2003/59934 A also contain examples of therapeutic polypeptides that may be fused to albumin or fragments thereof, and these examples apply also to the present invention.
  • Conjugates
  • The variants of albumin or fragments thereof according to the invention may be conjugated to a second molecule using techniques known within the art. Said second molecule may comprise a diagnostic moiety, and in this embodiment the conjugate may be useful as a diagnostic tool such as in imaging; or the second molecule may be a therapeutic compound and in this embodiment the conjugate may be used for therapeutic purposes where the conjugate will have the therapeutic properties of the therapeutic compound as well as the long plasma half-life of the albumin. Conjugates of albumin and a therapeutic molecule are known in the art and it has been verified that such conjugates have long plasma half-life compared with the non-conjugated, free therapeutic molecule as such. The conjugates may conveniently be linked via a free thio group present on the surface of HSA (amino acid residue 34 of mature HSA) using well known chemistry.
  • In one particular preferred aspect the variant albumin or fragment thereof is conjugated to a beneficial therapeutic compound and the conjugate is used for treatment of a condition in a patient in need thereof, which condition is responsive to the particular selected therapeutic compound. Techniques for conjugating such a therapeutically compound to the variant albumin or fragment thereof are known in the art. WO 2009/019314 discloses examples of techniques suitable for conjugating a therapeutically compound to a polypeptide which techniques can also be applied to the present invention. Further WO 2009/019314 discloses examples of compounds and moieties that may be conjugated to substituted transferrin and these examples may also be applied to the present invention. The teaching of WO 2009/019314 is included herein by reference.
  • HSA contains in its natural form one free thiol group that conveniently may be used for conjugation. As a particular embodiment within this aspect the variant albumin or fragment thereof may comprise further modifications provided to generate additional free thiol groups on the surface. This has the benefit that the payload of the variant albumin or fragment thereof is increased so that more than one molecule of the therapeutic compound can be conjugated to each molecule of variant albumin or fragment thereof, or two or more different therapeutic compounds may be conjugated to each molecule of variant albumin or fragment thereof, e.g., a compound having targeting properties such as an antibody specific for example a tumour; and a cytotoxic drug conjugated to the variant albumin or fragment thereof thereby creating a highly specific drug against a tumour. Teaching of particular residues that may be modified to provide for further free thiol groups on the surface can be found in copending patent application WO 2010/092135, which is incorporated by reference.
  • Associates
  • The variants of albumin or fragments thereof may further be used in form of โ€œassociatesโ€. In this connection the term โ€œassociateโ€ is intended to mean a compound comprising a variant of albumin or a fragment thereof and another compound bound or associated to the variant albumin or fragment thereof by non-covalent binding. As an example of such an associate can be mentioned an associate consisting variant albumin and a lipid associated to albumin by a hydrophobic interaction. Such associates are known in the art and they may be prepared using well known techniques. As an example of a preferred associate according to the invention can be mentioned an associate comprising variant albumin and paclitaxel.
  • Other Uses
  • The variant albumin or fragments thereof or fusion polypeptides comprising variant albumin or fragments thereof according to the invention have the benefit that their plasma half-life is altered compared to the parent albumin or fragments thereof or fusion polypeptides comprising parent albumin or fragments thereof. This has the advantage that the plasma half-life of conjugates comprising variant albumin or a fragment thereof or fusion polypeptide comprising variant albumin or a fragment thereof, or an associate comprising variant albumin or a fragment thereof according to the invention can be selected in accordance with the particular therapeutic purpose.
  • For example for a conjugate, associate or fusion polypeptide used for imaging purposes in animals or human beings, where the imaging moiety has an very short half-life and a conjugate or a fusion polypeptide comprising HSA has a plasma half-life that is far longer than needed for the imaging purposes it would be advantageous to use a variant albumin or fragment thereof of the invention having a shorter plasma half-life than the parent albumin or fragment thereof, to provide conjugates of fusion polypeptides having a plasma half-life that is sufficiently long for the imaging purpose but sufficiently short to be cleared form the body of the particular patient on which it is applied.
  • In another example for a conjugate, an associate or fusion polypeptide comprising a therapeutic compound effective to treat or alleviate a particular condition in a patient in need for such a treatment it would be advantageous to use the variant albumin or fragment thereof having a longer plasma half-life than the parent albumin or fragment thereof, to provide associates or conjugates or fusion polypeptides having longer plasma half-lives which would have the benefit that the administration of the associate or conjugate or fusion polypeptide of the invention would be needed less frequently or reduced dose with less side affects compared to the situation where the parent albumin or associates thereof or fragment thereof was used.
  • In a further aspect the invention relates to compositions comprising the variant albumin, associates thereof or fragment thereof, variant albumin fragment or associates thereof or fusion polypeptide comprising variant albumin or fragment thereof according to the invention. The compositions are preferably pharmaceutical compositions. The composition may be prepared using techniques known in the area such as disclosed in recognized handbooks within the pharmaceutical field.
  • In a particular embodiment the compositions comprise a variant albumin or a fragment thereof according to the invention and a compound comprising a pharmaceutically beneficial moiety and an albumin binding domain (ABD). According to the invention ABD means a site, moiety or domain capable of binding to circulating albumin in vivo and thereby conferring transport in the circulation of the ABD and any compound or moiety bound to said ABD. ABD's are known in the art and have been shown to bind very tight to albumin so a compound comprising an ABD bound to albumin will to a certain extent behave as a single molecule. The inventors have realized by using the variant albumin or fragment thereof according to the invention together with a compound comprising a pharmaceutically beneficial moiety and an ABD makes it possible to alter the plasma half-life of the compound comprising a pharmaceutically beneficial moiety and an ABD compared to the situation where said compound were injected as such in a patient having need thereof or administered in a formulation comprising natural albumin or a fragment thereof.
  • The variant albumin or fragments thereof, conjugates comprising variant albumin or a fragment thereof or fusion polypeptide comprising variant albumin or a fragment thereof, or an associate comprising variant albumin or a fragment thereof according to the invention may also be incorporated into nano- or microparticles using techniques well known within the art. A preferred method for preparing nano- or microparticles that may be applied to the variant albumins or fragments thereof according to the invention is disclosed in WO 2004/071536, which is incorporated herein by reference.
  • Compositions
  • The present invention is also directed to the use of a variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, or a conjugate comprising a variant of albumin or a fragment thereof, or an associate comprising a variant of albumin or a fragment thereof for the manufacture of a pharmaceutical composition, where in the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, or a conjugate comprising a variant of albumin or a fragment thereof, or an associate comprising a variant of albumin or a fragment thereof has an altered plasma half-life compared with HSA or the corresponding fragment thereof or fusion polypeptide comprising HSA or fragment thereof or conjugate comprising HSA.
  • In this connection the corresponding fragment of HSA is intended to mean a fragment of HSA that aligns with and has same number of amino acids as the fragment of the variant albumin with which it is compared. Similarly the corresponding fusion polypeptide comprising HSA or conjugate comprising HSA is intended to mean molecules having same size and amino acid sequence as the fusion polypeptide of conjugate comprising variant albumin, with which it is compared.
  • Preferably the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, or a conjugate comprising a variant of albumin or a fragment thereof has a plasma half-life that is higher than the plasma half-life of HSA or the corresponding fragment thereof or fusion polypeptide comprising HSA or fragment thereof.
  • Alternatively, this may be expressed as the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, or a conjugate comprising a variant of albumin or a fragment thereof has a KD to FcRn that is lower that the corresponding KD for HSA or the corresponding fragment thereof or fusion polypeptide comprising HSA or fragment thereof. Preferably, is KD for the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, or a conjugate comprising a variant of albumin or a fragment thereof less than 0.9ร—KD for HSA, more preferred less than 0.5ร—KD for HSA, more preferred less than 0.1ร—KD for HSA, even more preferred less than 0.05ร—KD for HSA, even more preferred less than 0.02ร—KD for HSA and most preferred less than 0.01ร—KD for HSA.
  • The variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, or a conjugate comprising a variant of albumin or a fragment thereof is preferably the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, or a conjugate comprising a variant of albumin or a fragment thereof according to the invention.
  • The present invention is further described by the following examples that should not be construed as limiting the scope of the invention.
  • Examples Materials and Methods ELISA:
  • Wells were coated with wild-type HSA or variants diluted in phosphate buffered saline (PBS) to stated concentrations, incubated overnight at 4 C and then blocked with 4% skimmed milk (Acumedia) for 1 hour at room temperature. The wells were then washed four times with PBS/0.005% TWEENยฎ 20 (PBS/T) pH 6.0 before glutathione-S-transferase (GST)-fused ( )shFcRn (0.5 ฮผg/ml) as described in FEBS J. 2008 August; 275(16):4097-110. pre-incubated with an horseradish peroxidase (HRP)-conjugated polyclonal anti-GST from goat (1:5000; GE Healthcare), diluted in 4% skimmed milk PBS/0.005% TWEENยฎ 20 (PBS/T) pH 6.0 was added to each well and incubated for 1.5 h at room temperature followed by washing four times with PBS/T pH 6.0. One hundred ฮผl of the substrate tetramethylbenzidine (TMB) (Calbiochem) was added to each well and incubated for 45 min before 100 ฮผl of 0.25 M HCl was added. The absorbance was measured at 450 nm using a Sunrise TECAN spectrophotometer (TECAN, Maennedorf, Switzerland).
  • The same ELISA was repeated with PBS/T pH 7.4.
  • Surface Plasmon Resonance (SPR):
  • SPR experiments were carried out using a Biacore 3000 instrument (GE Healthcare). Flow cells of CM5 sensor chips were coupled with shFcRn-GST (1400-5000 RU) using amine coupling chemistry as described in the protocol provided by the manufacturer. The coupling was performed by injecting 10 ฮผg/ml of the protein in 10 mM sodium acetate pH 5.0 (GE healthcare). Phosphate buffer (67 mM phosphate buffer, 0.15M NaCl, 0.005% TWEENยฎ 20) at pH 6.0) was used as running buffer and dilution buffer. Regeneration of the surfaces were achieved using injections of HBS-EP buffer (0.01M HEPES, 0.15M NaCl, 3 mM EDTA, 0.005% surfactant P20) at pH 7.4 (Biacore AB). For binding to immobilized shFcRn-GST, 1.0-0.5 ฮผM of each HSA variant was injected over the surface at constant flow rate (40 ฮผl/ml) at 25 C. In all experiments, data was zero adjusted and the reference cell subtracted. Data evaluation was performed using BIAevaluation 4.1 software (BIAcore AB).
  • The same SPR assay was repeated with HBS-EP buffer pH 7.4.
  • For the purposes of this patent unless otherwise stated HSA, WT HSA, rHA refer to Recombinant human serum albumin commercially available under the registered tradename RECOMBUMIN (available from Novozymes Biopharma UK Ltd, Nottingham UK) was used for the examples.
  • Serum albumin from other species: The albumins were recombinant whereas stated, produced using sequences provided from publicly available databases. Or purchased from commercial suppliers.
  • FcRn Expression and purification of soluble Human (shFcRn) and Mouse (smFcRn) FcRn: Methods for the generation of shFcRn and smFcRn expression plasmids, expression and purification of each heterodimer can be found in Berntzen et al. (2005) J. Immunol. Methods 298:93-104).Alternatively shFcRn FcRn heterodimer was produced by GeneArt AG (Germany). Sequences for the two sub units of the heterodimer can be found in SEQ ID NO: 3 and SEQ ID NO: 4. The soluble receptor was expressed in HEK293 cells and purified from culture supernatant using Ni-HiTrap chromatography columns.
  • Example 1. Preparation of Variants Preparation of Specific HSA Mutein Expression Plasmids
  • Methods for the expression of HSA mutant variants and HSA fusion variants were produced using several techniques. Standard molecular biology techniques were employed throughout such as described in Sambrook, J. and D. W. Russell, 2001. Molecular Cloning: a laboratory manual, 3rd ed. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.
  • Method 1. Amino Acid Substitutions in HSA Detailed in Table 1
  • Synthetic DNA NcoI/SacI fragments (859 bp) were generated by gene assembly (GeneArt AG, Germany) containing point mutations within the HSA-encoding gene (SEQ ID NO: 1) to introduce the desired amino acid substitution in the translated protein. Table 2 details the codons used to introduce the amino acid substitutions into the HSA-encoding gene. The nucleotide sequence of the synthetic fragment encoding unchanged amino acids (i.e. wild type) was identical to that in pDB2243 (described in WO 00/44772). The synthetic nucleotide fragments were ligated into NcoI/SacI-digested pDB2243 to produce plasmids pDB3876-pDB3886 (Table 1). For the production of expression plasmids, pDB3876-pDB3886 (see Table 1) were each digested with NotI and PvuI, the DNA fragments were separated through a 0.7% (w/v) TAE gel, and 2992 bp fragments (NotI cassettes' including PRB1 promoter, DNA encoding the fusion leader (FL) sequence (disclosed in WO 2010/092135), nucleotide sequence encoding HSA and ADH1 terminator; see FIG. 1) were purified from the agarose gel using a Qiagen Gel Extraction Kit following the manufacturer's instructions. โ€˜NotI cassettesโ€™ were ligated into a NotI/Shrimp Alkaline Phosphatase (Roche)-treated โ€œdisintegrationโ€ plasmid pSAC35, disclosed in EP-A-286 424 and described by Sleep, D., et al. (1991) Bio/Technology 9, 183-187. Ligation mixtures were used to transform chemically-competent E. coli DH5ฮฑ. Expression plasmids pDB3887-pDB3897, pSAC35-derivatives containing the โ€œNotI cassettesโ€, were identified using standard techniques. Disintegration plasmids pDB3887-pDB3897 and pDB2244 (For the expression of wild type HSA, described in WO 00/44772) (Table 1) were used to transform S. cerevisiae BXP10cir0 (as previously described WO/2001/079480 as described below.
  • TABLE 1
    Plasmid, amino acid substitution
    introduced into HSA
    Plasmid Construct
    pDB2244 HSA
    pDB3876 HSA D494N
    pDB3877 HSA D494A
    pDB3878 HSA E495Q
    pDB3879 HSA E495A
    pDB3880 HSA D494Q
    pDB3881 HSA D494N, T496A
    pDB3882 HSA T496A
    pDB3883 HSA E492G
    pDB3884 HSA E492G, V493P
    pDB3885 HSA E492P
    pDB3886 HSA E492H
    pDB3887 HSA D494N
    pDB3888 HSA D494A
    pDB3889 HSA E495Q
    pDB3890 HSA E495A
    pDB3891 HSA D494Q
    pDB3892 HSA D494N, T496A
    pDB3893 HSA T496A
    pDB3894 HSA E492G
    pDB3895 HSA E492G, V493P
    pDB3896 HSA E492P
    pDB3897 HSA E492H
    n/a = Not applicable.
    pDB3876-pDB3886 are sub-cloning plasmids.
  • TABLE 2
    Codons used to introduce amino
    acid substitutions into HSA
    Amino
    acid Codon
    Gly GGT
    Glu GAA
    Asp GAT
    Val GTT
    Ala GCT
    Arg AGA
    Lys AAA
    Asn AAT
    Met ATG
    Ile ATT
    Thr ACT
    Trp TGG
    Cys TGT
    Tyr TAT
    Leu TTG
    Phe TTT
    Ser TCT
    Gln CAA
    His CAT
    Pro CCA
    Stop TAA
  • Method 2. Production of HSA Variants D494N+E495Q+T496A and E495Q+T496A
  • A PCR-based method, using a QuickChange Lightening Kit (Statagene), was employed to introduce point mutations into HSA. Oligonucleotide pairs xAP094 (SEQ ID NO: 5)/xAP095 (SEQ ID NO: 6) and xAP096 (SEQ ID NO: 7)/xAP097 (SEQ ID NO: 8) were used to generate two HSA variants (D494N+E495Q+T496A and E495Q+T496A, respectively). Plasmid pDB3927 (disclosed in WO 2010/092135) was used as template DNA and the methodology recommended by the manufacturer of the kit was followed. The resulting plasmids were named pDB3995 and pDB3996 (contain HSA D494N+E495Q+T496A and E495Q+T496A expression cassettes, respectively). pDB3995 and pDB3996 were digested with BstEII/BsrBI and the linearised DNA molecules were purified using standard techniques. One hundred ng of each BstEII/BsrBI digested DNA, purified using a Qiagen PCR-Purification kit following the manufacturer's instructions, was mixed individually with 100 ng Acc65I/BamHI-digested pDB3936) (disclosed in WO 2010/092135) and used to directly transform S. cerevisiae BXP10cir0 using the Sigma Yeast Transformation kit described below.
  • Method 3. Amino Acid Substitutions in HSA Detailed in Table 3
  • Plasmid pDB3927 (disclosed in WO 2010/092135) (containing an identical nucleotide sequence encoding HSA as in pDB2243) was manipulated to amino acid substitutions within the mature HSA protein. Synthetic DNA fragments were generated (GeneArt AG, Germany or DNA2.0 Inc, USA) (NcoI/Bsu36I, AvrII/SphI or SacI/SphI fragments), containing point mutations within the HSA-encoding gene to introduce the desired amino acid substitution(s) into the translated protein sequence. Table 2 details the codons used to introduce the amino acid substitutions into the HSA-encoding gene. The nucleotide sequence of the synthetic fragment encoding unchanged amino acids (i.e. wild type) was identical to those in pDB3927. Synthetic DNA fragments were sub-cloned into NcoI/Bsu36I, AvrII/SphI-, SacI/Sph-digested pDB3927 (described in PCT 11527.204-WO) to generate pDB4006-pDB4010, pDB4083-pDB4101 and pDB4103-pDB4111 and pDB4194, pDB4200,pDB4202 (see Table 3).
  • Similarly, BamHI/SalI fragments containing point mutations in the nucleotide sequence encoding HSA were generated by gene assembly (DNA2.0 Inc, USA) and ligated into BamHI/SalI-digested pDB3964 (described in WO 2010/092135) to produce plasmids pDB3986-pDB3989 (Table 3).
  • The C-terminal string of amino acids from position 573-585 (KKLVAASQAALGL) (SEQ ID NO: 9) in HSA were mutated to those in macaque (PKFVAASQAALA) (SEQ ID NO: 10), mouse (PNLVTRCKDALA) (SEQ ID NO: 11), rabbit (PKLVESSKATLG) (SEQ ID NO: 12) and sheep (PKLVASTQAALA) (SEQ ID NO: 13) serum albumin. The codons used to introduce each amino acid substitution are given in Table 2. Synthetic DNA fragments (SacI/SphI) were generated (DNA2.0 Inc, USA) by gene assembly (the nucleotide sequence of the synthetic fragment encoding unchanged amino acids (i.e. wild type) was identical to that in pDB3927) and were sub-cloned into SacI/SphI-digested pDB3927 to produce plasmids pDB4114-4117 (Table 3).
  • Plasmids pDB3883 (Table 1), pDB4094 and pDB4095 (Table 3) were digested with NcoI/SacI and 857 bp fragments from each digest were purified before being ligated into NcoI/SacI-digested pDB4006 or pDB4110 (8.688 kb) (Table 3) to produce pDB4156-pDB4161.
  • Expression plasmids were generated in vivo (i.e. via homologous recombination in S. cerevisiae; a technique referred to as gap repair or in vivo cloningโ€”see Orr-Weaver & Szostak. 1983. Proc. Natl. Acad. Sci. USA. 80:4417-4421). Modified plasmids listed in Table 3 were digested with BstEII/BsrBI and the linearised DNA molecules were purified using standard techniques. One hundred ng of each BstEII/BsrBI digested DNA, purified using a Qiagen PCR-Purification kit following the manufacturer's instructions, was mixed individually with 100 ng Acc65I/BamHI-digested pDB3936 (disclosed in WO 2010/092135) and used to directly transform S. cerevisiae BXP10cir0 using the Sigma Yeast Transformation kit described below.
  • TABLE 3
    Plasmid Amino acid substitution in HSA
    pDB3986 HSA H440Q
    pDB3987 HSA H464Q
    pDB3988 HSA H510Q
    pDB3989 HSA H535Q
    pDB4006 HSA K573A
    pDB4007 HSA E492T/N503K/K541A
    pDB4008 HSA K541G
    pDB4009 HSA K541D
    pDB4010 HSA D550N
    pDB4083 HSA D494E/Q417H
    pDB4084 HSA Q417A
    pDB4085 HSA P499A
    pDB4086 HSA K500A
    pDB4087 HSA K536A
    pDB4088 HSA P537A
    pDB4089 HSA K538A
    pDB4090 HSA E492G/V493P/K538H/K541N/E542D
    pDB4091 HSA E492P/N503K/K541G/E542P
    pDB4092 HSA N503K
    pDB4093 HSA N503H
    pDB4094 HSA E492G/N503K
    pDB4095 HSA E492G/N503H
    pDB4096 HSA E492T
    pDB4097 HSA N503D
    pDB4098 HSA E492T/N503D
    pDB4099 HSA K538H
    pDB4100 HSA K541A
    pDB4101 HSA K541N
    pDB4103 HSA E542D
    pDB4104 HSA E542P
    pDB4105 HSA D550E
    pDB4106 HSA E492H/E501P/N503H/E505D/T506S/T540S/K541E
    pDB4107 HSA A490D/E492T/V493L/E501P/N503D/A504E/
    E505K/T506F/K541D
    pDB4108 HSA E501A
    pDB4109 HSA E501Q
    pDB4110 HSA K573P
    pDB4111 HSA E492G/K538H/K541N/E542D
    pDB4114 HSA K573P/L575F/G584A
    pDB4115 HSA K573P/K574N/A577T/A578R/S579C/Q580K/A581D/
    G584A
    pDB4116 HSA K573P/A577E/A578S/Q580K/A582T
    pDB4117 HSA K573P/A578S/S579T/G584A
    pDB4156 HSA E492G K573A
    pDB4157 HSA E492G N503K K573A
    pDB4158 HSA E492G N503H K573A
    pDB4159 HSA E492G K573P
    pDB4160 HSA E492G N503K K573P
    pDB4161 HSA E492G N503H K573P
    pDB4194 HSA D550E
    pDB4200 HSA K574N
    pDB4202 HSA Q580K
  • TABLEโ€ƒ4
    K500โ€ƒprimersโ€ƒandโ€ƒplasmids
    CODONS
    Originalโ€ƒprimers USED
    xAP216 CTTTGGAAGTCGACGAAACTTAC Gly GGT
    GTTCCAGGTGAATTCAACGCTG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ14)
    xAP217 CTTTGGAAGTCGACGAAACTTAC Glu GAA
    GTTCCAGAAGAATTCAACGCTG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ15)
    xAP218 CTTTGGAAGTCGACGAAACTTAC Asp GAC
    GTTCCAGACGAATTCAACGCTG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ16)
    xAP219 CTTTGGAAGTCGACGAAACTTAC Val GTT
    GTTCCAGTTGAATTCAACGCTG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ17)
    xAP220 CTTTGGAAGTCGACGAAACTTAC Arg AGA
    GTTCCAAGAGAATTCAACGCTG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ18)
    xAP221 CTTTGGAAGTCGACGAAACTTAC Asn AAC
    GTTCCAAACGAATTCAACGCTG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ19)
    xAP222 CTTTGGAAGTCGACGAAACTTAC Met ATG
    GTTCCAATGGAATTCAACGCTG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ20)
    xAP223 CTTTGGAAGTCGACGAAACTTAC Ile ATT
    GTTCCAATTGAATTCAACGCTG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ21)
    xAP224 CTTTGGAAGTCGACGAAACTTAC Thr ACC
    GTTCCAACCGAATTCAACGCTG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ22)
    xAP225 CTTTGGAAGTCGACGAAACTTAC Trp TGG
    GTTCCATGGGAATTCAACGCTG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ23)
    xAP226 CTTTGGAAGTCGACGAAACTTAC Cys TGT
    GTTCCATGTGAATTCAACGCTG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ24)
    xAP227 CTTTGGAAGTCGACGAAACTTAC Tyr TAC
    GTTCCATACGAATTCAACGCTG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ25)
    xAP228 CTTTGGAAGTCGACGAAACTTAC Leu TTG
    GTTCCATTGGAATTCAACGCTG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ26)
    xAP229 CTTTGGAAGTCGACGAAACTTAC Phe TTC
    GTTCCATTCGAATTCAACGCTG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ27)
    xAP230 CTTTGGAAGTCGACGAAACTTAC Ser TCT
    GTTCCATCTGAATTCAACGCTG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ28)
    xAP231 CTTTGGAAGTCGACGAAACTTAC Gln CAA
    GTTCCACAAGAATTCAACGCTG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ29)
    xAP232 CTTTGGAAGTCGACGAAACTTAC His CAC
    GTTCCACACGAATTCAACGCTG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ30)
    xAP233 CTTTGGAAGTCGACGAAACTTAC Pro CCA
    GTTCCACCAGAATTCAACGCTG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ31)
    xAP234 CTTTGGAAGTCGACGAAACTTAC STOP taa
    GTTCCATAAGAATTCAACGCTG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ32)
    xAP235 GAATT
    Figure US20180265570A1-20180920-P00001
    ATTACAAACCC
    AAAGCAGCTTGGGAAGC
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ33)
  • TABLEโ€ƒ5
    K573โ€ƒprimersโ€ƒandโ€ƒplasmids
    CODONS
    Originalโ€ƒprimers USED
    xAP187 ATAAGCCTAAGGCAGCTTGACTTGC Gly GGT
    AGCAACAAGTTTACCACCCTCCTCG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ34)
    xAP188 ATAAGCCTAAGGCAGCTTGACTTGC Glu GAA
    AGCAACAAGTTTTTCACCCTCCTCG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ35)
    xAP189 ATAAGCCTAAGGCAGCTTGACTTGC Asp GAT
    AGCAACAAGTTTATCACCCTCCTCG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ36)
    xAP190 ATAAGCCTAAGGCAGCTTGACTTGC Val GTT
    AGCAACAAGTTTAACACCCTCCTCG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ37)
    xAP191 ATAAGCCTAAGGCAGCTTGACTTGC Arg AGA
    AGCAACAAGTTTTCTACCCTCCTCG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ38)
    xAP192 ATAAGCCTAAGGCAGCTTGACTTGC Asn AAT
    AGCAACAAGTTTATTACCCTCCTCG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ39)
    xAP193 ATAAGCCTAAGGCAGCTTGACTTGC Met ATG
    AGCAACAAGTTTCATACCCTCCTCG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ40)
    xAP194 ATAAGCCTAAGGCAGCTTGACTTGC Ile ATT
    AGCAACAAGTTTAATACCCTCCTCG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ41)
    xAP195 ATAAGCCTAAGGCAGCTTGACTTGC Thr ACT
    AGCAACAAGTTTAGTACCCTCCTCG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ42)
    xAP196 ATAAGCCTAAGGCAGCTTGACTTGC Trp TGG
    AGCAACAAGTTTCCAACCCTCCTCG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ43)
    xAP197 ATAAGCCTAAGGCAGCTTGACTTGC Cys TGT
    AGCAACAAGTTTACAACCCTCCTCG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ44)
    xAP198 ATAAGCCTAAGGCAGCTTGACTTGC Tyr TAT
    AGCAACAAGTTTATAACCCTCCTCG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ45)
    xAP199 ATAAGCCTAAGGCAGCTTGACTTGC Leu TTG
    AGCAACAAGTTTCAAACCCTCCTCG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ46)
    xAP200 ATAAGCCTAAGGCAGCTTGACTTGC Phe TTT
    AGCAACAAGTTTAAAACCCTCCTCG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ47)
    xAP201 ATAAGCCTAAGGCAGCTTGACTTGC Ser TCT
    AGCAACAAGTTTAGAACCCTCCTCG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ48)
    xAP202 ATAAGCCTAAGGCAGCTTGACTTGC Gln CAA
    AGCAACAAGTTTTTGACCCTCCTCG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ49)
    xAP203 ATAAGCCTAAGGCAGCTTGACTTGC His CAT
    AGCAACAAGTTTATGACCCTCCTCG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ50)
    xAP204 ATAAGCCTAAGGCAGCTTGACTTGC STOP taa
    AGCAACAAGTTTTTAACCCTCCTCG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ51)
    xAP205 AATGCTG
    Figure US20180265570A1-20180920-P00002
    AGATCTGCTTGA
    ATGTGCTGATG
    (SEQโ€ƒIDโ€ƒNO:โ€ƒ52)
  • Method 4. HSA K500 and K573 Permutation Library
  • PCR was used to produce two permutation libraries in which the codons encoding amino acid 500 or 573 of mature HSA were changed (mutated) to alternative non-wild type amino acids and a termination codons (K5XXSTOP). Mutagenic oligonucleotides (Table 4 and Table 5), were designed to amplify HSA-encoding DNA and incorporate the desired changes. That is, for the changes at position 500, pDB4082 (FIG. 1) was used as a template DNA. pDB4082 is a derivative of pDB2305 (disclosed in EP1788084) and was produced as follows. pDB2305 (FIG. 2) was digested with NsiI/SpeI and the yielded 8.779 kb NsiI fragment was self-ligated to produce pDB4005 (FIG. 3). A synthetic DNA fragment (BsaI/SphI) was generated by gene assembly (DNA2.0 Inc, USA) (SEQ ID NO: 1) (containing 3โ€ฒ region of the PRB1 promoter, modified fusion leader sequence, nucleotide sequence encoding HSA and 5โ€ฒ region of the modified ADH1 terminator), and ligated into HindIII/SphI-digested pDB4005 (FIG. 3) to produce pDB4082. Note. The HindIII site in PRB1 promoter site has been removed and a SacII site within the nucleotide sequence encoding HSA has been introduced.
  • For the permutation library for position 500 of HSA, the nucleotide sequence encoding HSA corresponding to that between the Sail/HindIII sites (see plasmid map pDB4082, FIG. 1) was generated using the New England Biolabs Phusion kit (Table 6) and oligonucleotides listed in Table 4. Table 7 describes the PCR method employed.
  • The permutation library at amino acid position 573 in HSA was generated using pDB3927 as template DNA and involved amplifying the albumin-encoding DNA corresponding to that between the NcoI and Bsu36I sites using oligonucleotides detailed in Table 5.
  • TABLE 6
    PCR ingredients
    500 library 573 library
    20โ–กI Buffer HF (5X)
    2โ–กI dNTP mix (10 mM)
    2โ–กI oligonucleotide xAP235 xAP205
    (10โ–กM)
    2โ–กI oligonucleotide xAP216- xAP187-
    (10โ–กM) xAP234 xAP204
    1โ–กI Phusion polymerase
    1โ–กI Template DNA pDB4082 pDB3927
    (~5 ng)
    72โ–กI dH2O
  • TABLE 7
    PCR conditions:
    98ยฐ C. for 2 min โ€‚1 cycle
    98ยฐ C. for 10 sec 35 cycles
    57ยฐ C. for 30 sec
    72ยฐ C. for 20 sec
    72ยฐ C. for 5 min โ€‚1 cycle
  • For the albumin variants based at positions 500 and 573, each PCR-product was purified using a Qiagen PCR-clean up kit (according to the manufactures instructions), digested with SalI/HindIII (position 500 library) or NcoI/Bsu36I (position 573 library). The digested DNAs were then purified using a Qiagen PCR-clean up kit and ligated into SalI/HindIII- or NcoI/Bsu36I-digested pDB4082 or pDB3927, respectively, replacing the equivalent native sequence. Ligations were transformed into E. coli DH5ฮฑ, subsequent plasmids isolated from transformants using a Qiagen miniprep kit (according to the manufacturer's instructions) and the correct constructs identified by restriction analysis. This produced a collection of plasmids, pDB4204-pDB4222 (position 500 library) pDB4173 to pDB4190 (position 573 library), containing albumin genes which differed only in their sequence corresponding to the codon for the amino acid at position 500 or 573 Table 4 and 5, respectively). The specific changes in each plasmid were confirmed by sequencing.
  • The resultants plasmids were used to generate expression plasmids and albumin fusion producing yeast by in vivo cloning as described above. That is, S. cerevisiae was transformed using the Sigma Yeast Transformation kit (described below), using a mixture of a 100 ng BstEII/BsrBI-digeste HSA variant containing plasmid and 100 ng Acc65I/BamHI digested pDB3936.
  • Transformation of S. cerevisiae
  • S. cerevisiae BXP10 cir0 (as previously described WO/2001/079480) or Strain A cir0 (described in WO/2005/061718) was streaked on to YEPD plates (1% (w/v) yeast extract, 2% (w/v) Bactopeptone, 2% (w/v) glucose), 1.5% agar) and allowed to grow for 4 days at 30ยฐ C. prior to transformation. One ฮผg of whole plasmid (i.e. circular plasmids) or, for gap repair, 100 ng BstEII/BsrBI- or NsiI/PvuI-digested HSA variant or HSA variant fusion containing plasmid and 100 ng Acc65I/BamHI digested pDB3936 were used to transform S. cerevisiae using a Sigma Yeast Transformation kit using a modified lithium acetate method (Sigma yeast transformation kit, YEAST-1, protocol 2; Ito et al. (1983) J. Bacteriol., 153, 16; Elble, (1992) Biotechniques, 13, 18). The protocol was amended slightly by incubating the transformation at room temperature for 4 h prior to heat shock. Following heat shock, the cells were briefly centrifuged before being re-suspended in 200 ฮผl 1M sorbitol then spread over BMMD agar plates, the composition of BMMD is described by Sleep et al., (2001), Yeast, 18, 403. Plates were incubated at 30ยฐ C. for 4 days before individual colonies were patched on to fresh BMMD plates. Yeast strain numbers are detailed in Table 1.
  • Stocks were prepared for each yeast strain as follows: BMMD broth was inoculated with a heavy loop of each yeast patch and grown for 24 h at 30ยฐ C. with orbital shaking at 200 rpm. Cells were harvested by centrifugation at 1900ร—g for 5 min in a Sorval RT600 centrifuge, 15 mL supernatant was removed and replaced by trehalose 40% (w/v). The cells were resuspended and transferred to cyrovials (1 mL) for storage at โˆ’80ยฐ C.
  • Shake Flask Growth of S. cerevisiae
  • BMMD (recipe 0.17% (w/v) yeast nitrogen base without amino acid and ammonium sulphate (Difco), 37.8 mM ammonium sulphate, 29 mM citric acid, 142 mM disodium hydrogen orthophosphate dehydrate pH6.5, 2% (w/v) glucose) media (10 mL) was inoculated with each yeast strain and grown for 12 h at 30ยฐ C. with orbital shaking at 200 rpm. An aliquot of each starter culture (4 mL) was used to inoculate 2ร—200 mL BMMD media and grown for 36 h at 30ยฐ C. with orbital shaking at 200 rpm. Cells were harvested by filtration through 0.2 ฮผm vacuum filter membranes (Stericup, Millipore) including a GF-D prefilter (Whatman) and the supernatant retained for purification.
  • Primary Concentration
  • Retained culture supernatant was concentrated using Tangential Flow Filtration using a Pall Filtron LV system fitted with a Omega 10KD (0.093 sqยทm2) filter (LV Centramateโ„ข cassette, Pall Filtron) with a transmembrane pressure of 20 psi and a recirculation rate of 180 mLยทminโˆ’1.
  • Fermentation
  • Fed-batch fermentations were carried out in a 10 L Sartorius Biostat C fermenter at 30ยฐ C.; pH was monitored and adjusted by the addition of ammonia or sulphuric acid as appropriate. The ammonia also provided the nitrogen source for the cultures. The level of dissolved oxygen was monitored and linked to the stirrer speed, to maintain the level at >20% of saturation. Inocula were grown in shake flasks in buffered minimal media (recipe). For the batch-phase the cultures was inoculated into fermenter media (approximately 50% of the fermenter volume) containing 2% (w/v) sucrose. The feed stage was automatically triggered by a sharp rise in the level of dissolved oxygen. Sucrose was kept at growth-limiting concentrations by controlling the rate of feed to a set nominal growth rate. The feed consisted of fermentation media containing 50% (w/v) sucrose, all essentially as described by Collins. (Collins, S. H., (1990) Production of secreted proteins in yeast, in: T. J. R. Harris (Ed.) Protein production by biotechnology, Elsevier, London, pp. 61-77).
  • GP-HPLC Quantitation
  • Purified albumin variants, fusions and conjugates were analysed by GP-HPLC and quantification as follows. Injections of 25 ฮผL were made onto a 7.8 mm idร—300 mm length TSK G3000SWXL column (Tosoh Bioscience), with a 6.0 mm idร—40 mm length TSK SW guard column (Tosoh Bioscience). Samples were chromatographed in 25 mM sodium phosphate, 100 mM sodium sulphate, 0.05% (w/v) sodium azide, pH 7.0 at 1 mL/min, Samples were quantified by UV detection at 280 nm, by peak area, relative to a recombinant human albumin standard of known concentration (10 mg/mL) and corrected for their relative extinction coefficients.
  • Purification of Albumin Variants from Shake Flask
  • Albumin variants were purified from shake flask (either culture supernatant or concentrated culture supernatant) using a single chromatographic step using an albumin affinity matrix (AlbuPureโ„ขโ€”ProMetic BioSciences, Inc.). Chromatography was performed at a constant linear velocity of 240 cm/h throughout. Culture supernatant was applied to a 6 cm bed height, 2.0 mL packed bed pre-equilibrated with 50 mM sodium acetate pH5.3. Following load the column was washed with 10 column volume (CV) of equilibration buffer, then 50 mM ammonium acetate pH8.0 (10CV). Product was eluted with either 50 mM ammonium acetate 10 mM octanoate pH8.0, 50 mM Ammonium Acetate 30 mM Sodium Octanoate 200 mM Sodium Chloride pH7.0 or 200 mM Potassium thiocyanate. The column was cleaned with 0.5M NaOH (3cv) and 20 mM NaOH (3.5cv). Eluate fraction from each albumin variant were concentrated and diafiltered against 10 volumes of 50 mM sodium chloride (Vivaspin20 10,000 MWCO PES with optional diafiltration cups, Sartorius). Purified albumin variants were quantified by GP-HPLC as described above.
  • Purification of Albumin-Fusion Variants from Shake Flask
  • Albumin-fusion variants were purified from shake flask culture supernatant using a single chromatographic step using an albumin affinity matrix (AlbuPureโ„ขโ€”ProMetic BioSciences, Inc.). Chromatography was performed at a constant linear velocity of 240 cm/h throughout. Culture supernatant or concentrated culture supernatant was applied to a 6 cm bed height, 2.0 mL packed bed pre-equilibrated with 50 mM sodium acetate pH5.3. Following load the column was washed with 10 column volume (cv) equilibration buffer then 50 mM ammonium acetate pH8.0 (10cv). Product was eluted with either 50 mM ammonium acetate 10 mM octanoate pH8.0, 50 mM Ammonium Acetate 30 mM Sodium Octanoate 200 mM Sodium Chloride pH7.0, 50 mM Ammonium Acetate 100 mM Sodium Octanoate pH9.0 or 200 mM Potassium thiocyanate. The column was cleaned with 0.5M NaOH (3cv) and 20 mM NaOH (3.5cv). Eluate fraction from each albumin variant-fusion were concentrated and diafiltered against 10 volumes of 25 mM Tris, 150 mM NaCl, 2 mM KCl, pH 7.4 (Vivaspin20 10,000 MWCO PES with optional diafiltration cups, Sartorius). Purified albumin-fusion variants were quantified by GP-HPLC as described above.
  • Purification of Albumin Variants from Fermentation
  • Albumin variants were purified from high cell density fed batch fermentation supernatants after separation by centrifugation, using a Sorvall RC 3C centrifuge (DuPont). Culture supernatant was chromatographed through an 11 cm bed height column 8.6 mL packed bed packed with a custom synthesised albumin affinity matrix (AlbuPureโ„ขโ€”ProMetic BioSciences, Inc.) as described above. Product was eluted using elution buffers describe above at a flow rate of 120 cm/h. The eluate fraction(s) was analysed by GP-HPLC. (above).and reducing SDS-PAGE for purity and if required concentrated (Vivaspin20 10,000 MWCO PES) and applied to a 2.4ร—96 cm column packed with Superdex 75 run at a flow rate of 39 cm/h in 25 mM Tris, 150 mM NaCl, 2 mM KCl, pH 7.4. The peak was fractionated, assayed by GP-HPLC and pooled in order to generate the monomeric protein of interest. Pooled fractions were concentrated (Vivaspin20 10,000 MWCO PES, Sartorius). All proteins to be assayed for receptor (FcRn) binding properties and or other analysis were quantified by GP-HPLC as described above corrected for their relative extinction coefficients.
  • Example 2. Determination of Receptor (shFcRn) Binding Properties of Blood Derived HSA and Recombinant Human Albumin
  • Essentially fatty acid-free HSA (Sigma-Aldrich) was further purified by size exclusion chromatography as described in Andersen et al (2010). J. Biol. Chem. 285, (7), 4826-4836. Ten ฮผM of monomeric HSA and rHA were analysed using SPR as described above and the data presented in FIG. 4.
  • Direct comparison of HSA (blood derived) with recombinant human albumin (Recombumin) at the same concentration (10 ฮผM) (FIGS. 4A and 4B) shows for both samples binding to immobilized shFcRn (pH6.0, pH7.4 respectively) was reversible and pH dependent. In addition, comparison of HSA vs recombinant human albumin by Bosse et al (2005). J. Clin. Pharmacol. 45; 57-67, demonstrated equivalent half life in vivo human study
  • Example 3. Determination of Receptor (shFcRn) Binding Properties of Albumin Variants
  • Two established FcRn binding assays were used, ELISA and SPR. There are major differences between the assays: In the ELISA system HSA is coated directly in wells and shFcRn-GST is added in solution whereas in the SPR assay shFcRn-GST is immobilized to a CM5 chip and HSA injected in solution. The pH can be varied in both systems.
  • The variants were analysed using ELISA at pH 6.0 and pH 7.4. Results are disclosed in FIG. 5. The ELISA values represent the mean of duplicates.
  • The variants were analysed using SPR analysis at pH 6.0 and pH 7.4. Results are disclosed for a representative number of variants in FIG. 6 using a concentration of the variants of 0.2 ฮผM and in FIG. 7 using a concentration of the variants of 1 ฮผM.
  • The SPR data disclosed in FIGS. 6 and 7 were normalized and the relative binding of variants at each concentration is shown in FIGS. 8 A and B respectively.
  • The conclusions of the analysis are that all tested variants have the characteristic binding to the receptor at pH 6.0 but no binding at pH 7.4. The variants D494N,Q,A, E495Q,A, T496A, and D494N+T496A show reduced binding to the receptor compared to HSA.
  • Example 4. Determination of Receptor (shFcRn/smFcRn) Binding Properties of Albumin Variants
  • Using the SPR analysis method below the association constant Ka, the dissociation constant Kd and the binding constant KD calculated for HSA and mouse serum albumin (MSA) binding to human and mouse FcRn (Table 8).
  • SPR Analysesโ€”
  • SPR analyses were performed on a BIAcore 3000 instrument (GE Healthcare) using CM5 chips and immobilization of smFcRn-GST and shFcRn-GST variants or smFcRn was performed using the amine coupling kit (GE Healthcare). Protein samples (10 ฮผg/ml) were injected in 10 mM sodium acetate at pH 4.5 (GE Healthcare), all as described by the manufacturer. Unreacted moieties on the surface were blocked with 1 M ethanolamine. For all experiments, phosphate buffer (67 mM phosphate buffer, 0.15 M NaCl, 0.005% TWEENยฎ 20) at pH 6.0 or pH 7.4, or HBS-P buffer (0.01 M HEPES, 0.15 M NaCl, 0.005% surfactant P20) at pH 7.4 were used as running buffer or dilution buffer. Kinetic measurements were performed using a low density immobilized surface (100-200 resonance units (RU)). Serial dilutions of hIgG1 (2000.0-31.2 nM), mIgG1 (1000.0-15.6 nM), MSA (20.0-0.3 ฮผM) and HSA (200.0-3.1 ฮผM) were injected at pH 6.0 or pH 7.4, at a flow rate 50 ฮผI/minute at 25ยฐ C. Additive binding was recorded by injecting HSA (10 ฮผM), MSA (5 ฮผM), hIgG1 (100 nM) or mIgG1 (100 nM) alone or two at a time at 25ยฐ C. at 20 ฮผI/minute at pH 6.0 over immobilized shFcRn (หœ600 RU) or smFcRn (หœ600 RU). Competitive binding was measured by injecting shFcRn (50 nM) or smFcRn (100 nM) alone or together with different amounts of HSA or MSA (10.0-0.05 ฮผM) over immobilized HSA (หœ2600 RU) or MSA (หœ2000 RU). In all cases, to correct for nonspecific binding and bulk buffer effects, responses obtained from the control surfaces and blank injections were subtracted from each interaction curve. Kinetic rate values were calculated using predefined models (Langmuir 1:1 ligand model, heterogeneous ligand model and steady state affinity model) provided by the BIAevaluation 4.1 software. The closeness of the fit, described by the statistical value ฯ‡2 that represents the mean square, was lower than 2.0 in all affinity estimations.
  • TABLE 8
    Binding constants of HSA and MSA shFcRn and smFcRn.
    KD
    Albumin FcRn Ka Kd KD Req.
    Species Species (103/Ms) (103/s) (ฮผM) (ฮผM)
    MSA Mouse 4.2 ยฑ 0.5 39.4 ยฑ 3.1 9.3 ยฑ 0.4 NDd
    MSA Human 3.8 ยฑ 0.0 โ€‚3.1 ยฑ 0.1 0.8 ยฑ 0.2 NDโ€‚
    HSA Mouse NA NA NA 86.2 ยฑ 4.1
    HSA Human 2.7 ยฑ 1.3 12.2 ยฑ 5.9 4.5 ยฑ 0.1 โ€‚4.6 ยฑ 0.5
  • The KD's were generated using the BIAevaluation 4.1 software) A Langmuir 1:1 ligand model was used throughout. The kinetic values represent the average of triplicates. ND means: Not determined. NA means: Not acquired
  • Example 5. Binding of Albumins from Other Species to Human FcRn
  • Commercially available animal albumin (either Sigma-Aldrich or Calbiochem) were further purified as described in Andersen et al (2010). J. Biol. Chem. 285, (7), 4826-4836. The binding of donkey serum albumin, bovine serum albumin, goat serum albumin, sheep serum albumin, rabbit serum albumin, dog serum albumin, hamster serum albumin, guinea pig albumin, rat serum albumin and chicken serum albumin to shFcRn was determined using the techniques described in Materials and Methods. The ELISA results are disclosed in FIG. 9 A-D and the relative bindings summarized in FIG. 9 E.
  • The SPR results are shown in FIG. 10, where the binding at pH 6.0 and pH 7.4 for each albumin species are shown. Table 10 shows an overview of the relative binding responses measured using ELISA and SPR:
  • TABLE 10
    Cross-species albumin-FcRn binding
    shFcRn
    Albumin ELISA SPR
    specie pH6.0 pH7.4 pH6.0 pH7.4
    Human ++(+) โˆ’ ++(+) โˆ’
    Donkey +++ โˆ’ ++ โˆ’
    Cow ++ โˆ’ ++ โˆ’
    Sheep +/โˆ’ โˆ’ โˆ’ โˆ’
    Goat +/โˆ’ โˆ’ โˆ’ โˆ’
    Rabbit ++++ โˆ’ +++ โˆ’
    Dog NDa ND +++ โˆ’
    G. pig ++++ + ++++ +
    Hamster +++ โˆ’ +++ โˆ’
    Rat +++ โˆ’ +++ โˆ’
    Mouse +++ โˆ’ +++ โˆ’
    Chicken โˆ’ โˆ’ โˆ’ โˆ’

    Relative binding responses are categorized from strongest (++++) to weakest (+) and no binding (โˆ’). a: Not determined (ND).
  • A hierarchy ranging from strongest to weakest binding is as follows; guinea pig=1>rabbit>hamster/dog>rat/mouse>donkey>human>bovine>goat/sheep>chicken. This data shows that animal albumins have different affinities for shFcRn.
  • Example 6. Kinetics of the HSA Variant for shFcRn
  • The binding constants for variants according to the invention were determined according to the methods described in Materials and Methods.
  • TABLE 11
    Binding constants of HSA variants for shFcRn
    KD
    Albumin Ka kd KD Req
    Variant (103/Ms) (10โˆ’3/s) (ฮผM) (ฮผM)
    WT 3.2 ยฑ 0.2 15.5 ยฑ 2.5 4.8 5.4
    D494N 1.7 ยฑ 0.0 18.6 ยฑ 0.0 10.9 11.8
    D494A 2.3 ยฑ 0.1 53.4 ยฑ 0.3 23.2 17.0
    D494Q 2.1 ยฑ 0.0 58.2 ยฑ 3.8 27.7 ND
    E495Q 2.5 ยฑ 0.0 24.1 ยฑ 0.2 9.6 10.9
    E495A 2.1 ยฑ 0.0 14.0 ยฑ 0.0 7.0 8.6
    D494N + T496A 2.5 ยฑ 0.0 11.0 ยฑ 0.0 4.4 5.5
    T496A 2.3 ยฑ 0.0 11.7 ยฑ 0.5 5.1 7.1
    E492G 4.1 ยฑ 0.0 11.0 ยฑ 0.0 2.7 ND
  • The KD's were generated using the BIAevaluation 4.1 software) A Langmuir 1:1 ligand model was used throughout. The kinetic values represent the average of triplicates. ND means: Not determined.
  • The results correspond with the conclusions made in Example 3 based on SPR and ELISA data but in addition shows that E492G has increased affinity to its receptor,
  • Example 7. Competitive Analysis of the HSA Variants
  • Competitive analysis of the HSA variants prepared in example 1 and WT HSA was performed using the methods described in example 4. Results are shown in FIG. 15. The results show that the variant E492G, unlike E492H E492P and E492G+V493P, has stronger binding to shFcRn than HSA.
  • Example 8. Analysis of Q417 Substitutions
  • Using the method of Example 1 variants of HSA having the substitutions Q417A and D494E+Q417H were constructed. The kinetic properties of these variants were tested using the methods in Materials and Methods and are shown in Table 12.
  • TABLE 12
    Binding constants of HSA variants for shFcRn
    KD
    ka kd KDb Reqc
    Albumin varianta (103/Ms) (10โˆ’3/s) (ฮผM) (ฮผM)
    WT 3.2 ยฑ 0.2 15.5 ยฑ 2.5 4.8 5.4
    Q417A 3.2 ยฑ 0.1 26.0 ยฑ 0.0 8.1 ND
    D494E + Q417H 3.1 ยฑ 0.1 20.5 ยฑ 0.5 6.6 ND
    aDilutions of HSA variants were injected over immobilized shFcRn (~1500 RU).
    bThe kinetic rate constants were obtained using a simple first-order (1:1) bimolecular interaction model.
    cThe steady state affinity constant was obtained using an equilibrium (Req) binding model supplied by the BIAevaluation 4.1 software. The kinetic values represent the average of triplicates.
    d: Not determined (ND).
  • The data show that variants Q417A and D494E+Q417H bind weaker to the receptor than the wild-type HSA.
  • Example 9. Analysis of HSA Variants in Position 499, 500, 536, 537, 538 and 573
  • Using the method of Example 1 variants of HSA having the substitutions P499A, K500A, K536A, P537A, K538A and K573A were constructed. The receptor binding properties of these variants were tested as described in Materials and Methods. Results are shown in FIG. 11.
  • The data demonstrated that variants P499A, K536A, P537A and K538A had a reduced binding affinity to shFcRn relative to HSA. Variant K500A had almost completely lost its ability to bind to shFcRn and K573A had an increased binding affinity to shFcRn both relative to HSA.
  • Example 10. Analysis of Variants in Position 501 of HSA
  • Using the method of Example 1 variants of HSA having the substitutions E501A and E501Q were constructed. The kinetic properties of these variants were tested as described in Materials and Methods.
  • TABLE 13
    Binding constants of HSA variants for shFcR
    KD
    ka kd KDb Reqc
    Albumin varianta (103/Ms) (10โˆ’3/s) (ฮผM) (ฮผM)
    WT 3.2 ยฑ 0.2 15.5 ยฑ 2.5 4.8 5.4
    E501A 3.3 ยฑ 0.0 26.0 ยฑ 0.0 7.8 ND
    E501Q 2.7 ยฑ 0.1 15.5 ยฑ 0.5 5.7 ND
    aDilutions of HSA variants were injected over immobilized shFcRn (~1500 RU).
    bThe kinetic rate constants were obtained using a simple first-order (1:1) bimolecular interaction model.
    cThe steady state affinity constant was obtained using an equilibrium (Req) binding model supplied by the BIAevaluation 4.1 software. The kinetic values represent the average of triplicates
    d: Not determined (ND).
  • The data shows that variants E501A and E501Q have a slightly decreased binding affinity to shFcRn relative to HSA.
  • Example 11. Analysis of HSA Variants in Position 573
  • Using the method of Example 1 variants of HSA having a substitution at position 573 were constructed. All variants at position 573 were generated and the receptor binding properties of these variants were tested as described in Materials and Methods but with SPR analysis performed at pH5.5. Results are shown in the table 14 below and FIGS. 12 and 13.
  • TABLE 14
    Kinetics of HSA K573 single point mutants.
    ka kd KDb
    Albumin varianta (103/Ms) (10โˆ’3/s) (nM)
    WT 9.0 ยฑ 0.0 6.9 ยฑ 0.1 766
    K573A 7.4 ยฑ 0.0 2.2 ยฑ 0.0 297
    K573C 4.2 ยฑ 0.0 1.1 ยฑ 0.2 262
    K573D 7.9 ยฑ 0.2 4.1 ยฑ 0.3 518
    K573E 9.0 ยฑ 0.0 2.9 ยฑ 0.0 322
    K573F 7.8 ยฑ 0.1 0.5 ยฑ 0.1 74
    K573G 8.5 ยฑ 0.0 1.8 ยฑ 0.1 212
    K573H 12.0 ยฑ 0.2โ€‚ 0.8 ยฑ 0.0 68
    K573I 8.6 ยฑ 0.0 0.8 ยฑ 0.2 99
    K573L 5.1 ยฑ 0.2 2.3 ยฑ 0.1 451
    K573M 8.6 ยฑ 0.0 1.9 ยฑ 0.0 221
    K573N 7.3 ยฑ 0.2 1.1 ยฑ 0.3 151
    K573P 9.8 ยฑ 0.0 0.6 ยฑ 0.1 61
    K573Q 7.7 ยฑ 0.2 2.6 ยฑ 0.0 338
    K573R 8.5 ยฑ 0.0 3.0 ยฑ 0.2 353
    K573S 7.9 ยฑ 0.2 1.2 ยฑ 0.2 152
    K573T 8.7 ยฑ 0.2 1.1 ยฑ 0.1 126
    K573V 8.1 ยฑ 0.0 0.6 ยฑ 0.2 80
    K573W 15.0 ยฑ 0.2โ€‚ 0.4 ยฑ 0.3 29
    K573Y 22.0 ยฑ 0.1โ€‚ 0.5 ยฑ 0.1 23
    K573STOP ND ND 141000
    aDilutions of HSA variants were injected over immobilized shFcRn (~1500 RU).
    bThe kinetic rate constants were obtained using a simple first-order (1:1) bimolecular interaction model.
    cThe steady state affinity constant was obtained using an equilibrium (Req) binding model supplied by the BIAevaluation 4.1 software. The kinetic values represent the average of duplicates.
    d: Not determined (ND).
  • The results show that all variants having substitution in position 573 have improved binding to shFcRn compared with WT HSA. In particular the variants K573F, K573H, K573P, K573W and K573Y have more than 10 fold lower KD to shFcRn than the parent HSA. The variant K573STOP is a truncated albumin having a stop codon in position 573. The sensorgram for the K573STOP variant show significantly reduced binding compare to the WT HSA and generated a high KD. The increased affinity that we have shown for the variant K573E, a natural variant characterized by Otagiri (2009). Biol. Pharm. Bull. 32(4) 527-534, is predicted to have increased half-life in vivo.
  • Example 12. Analysis of Further HSA Variants
  • Using the method of Example 1 variants of HSA having the substitutions E492G, E492G+N503H, N503H, D550E, E492G+N503K, E542P, H440Q, K541G, K541D, D550N E492G+K538H+K541N+E542D, E492T+N503K+K541A, E492P+N503K+K541G+E542P, E492H+E501P+N503H+E505D+T506S+T540S+K541E, A490D+E492T+V493L+E501P+N503D+A504E+E505K+T506F+K541D, E492G+V493P+K538H+K541N+E542D were constructed. The receptor binding properties of these variants were tested as described in Materials and Methods, and the results are shown in Table 15 and FIG. 14.
  • TABLE 15
    Binding constants of HSA variants for shFcR
    KD
    Albumin Ka kd KDb Reqc
    varianta (103/Ms) (10โˆ’3/s) (ฮผM) (ฮผM)
    WT 3.2 ยฑ 0.2 15.5 ยฑ 2.5 4.8 5.4
    E492G 4.1 ยฑ 0.0 11.0 ยฑ 0.0 2.7 ND
    E492G/N503H 6.9 ยฑ 0.1 14.5 ยฑ 0.5 2.1 ND
    N503H 5.4 ยฑ 0.0 24.0 ยฑ 0.1 4.4 ND
    D550E 3.2 ยฑ 0.4 11.8 ยฑ 0.0 3.6 ND
    E492G/N503K 5.9 ยฑ 0.1 16.0 ยฑ 0.0 2.7 ND
    E542P 3.4 ยฑ 0.0 15.7 ยฑ 0.2 4.7 ND
    H440Q 3.2 ยฑ 0.1 20.8 ยฑ 0.0 6.5 ND
    K541G 3.2 ยฑ 0.0 23.0 ยฑ 0.0 7.1 ND
    K541D 2.6 ยฑ 0.0 24.0 ยฑ 0.0 9.2 ND
    D550N 2.5 ยฑ 0.0 30.0 ยฑ 0.0 12.0 ND
    aDilutions of HSA variants were injected over immobilized shFcRn (~1500 RU).
    bThe kinetic rate constants were obtained using a simple first-order (1:1) bimolecular interaction model.
    cThe steady state affinity constant was obtained using an equilibrium (Req) binding model supplied by the BIAevaluation 4.1 software. The kinetic values represent the average of triplicates.
    d: Not determined (ND).
  • The results show that for position 550, a substitution to E results in an increased affinity whilst a substitution to N resulted in reduced affinity for shFcRn at pH6.0. When this analysis was repeated for the D550E substitution at pH5.5 however no observable increase in affinity was seen. The substituted for an acid amino acid (E) maintains and improves the binding. However the substitution for an uncharged amide amino acid reduces binding at pH6.0. Based on this observation, we would predict for this position that substitutions to basic amino acids (H, K and R) would result in further reductions in binding.
  • Example 13. Mutations in His Residues
  • The following variants were generated using the methods described in Example 1: H440Q, H464Q, H510Q and H535Q. FIG. 15 shows SPR sensorgrams of these variants interacting with shFcRn as described in Materials and Methods.
  • It was found that the variant H440Q bound with comparable affinity as HSA. In contrast H464Q, H510Q and H535Q had significantly reduced affinity to shFcRn. This supports the previously published observations that mutagenesis of these Histidine residues significantly reduced HSA binding to shFcRn (Wu et al (2010). PEDS, 23(10)789-798). Wu et al show a reduced half-life for a diabody fusion proteins (scFv-DIII)2 in mice with an order of removal from slowest to fastest: Db-DIII WT>H535A>H510A>H464A>Db. Based on affinity to shFcRn and when compared to smFcRn (example 5) we would predict the clearance order in humans to be (for glutamine (Q) substitutions) WT>H440Q>H510Q>H464Q>H535Q.
  • Example 14. Further Variants
  • The following variants were generated using the methods described in Example 1: K574N and Q580K in HSA. Binding of the variants to FcRn was tested using the SPR assay as described in Materials and Methods and the results are shown in Table 16.
  • The results show that variants K574N and Q580K bound stronger to shFcRn.
  • TABLE 16
    Following kinetic data was found for these variants:
    ka kd KD
    Albumin variant (103/Ms) (10โˆ’3/s) (ฮผM)
    WT 9.7 ยฑ 0.0 30.0 ยฑ 0.1 3.1
    K574N 4.9 ยฑ 01โ€‰ โ€‚8.4 ยฑ 0.1 1.7
    Q580K 6.0 ยฑ 0.0 โ€‚9.3 ยฑ 0.0 1.5
  • Example 15. Analysis of HSA Variants in Position 500
  • Using the method of Example 1 variants of HSA having a substitution at position 500 were constructed. All variants at position 500 were generated and the receptor binding properties of these variants were tested. Biacore X, Biacore X100 and Sensor Chip CM5 were used for all analyses, both supplied by G E Healthcare. shFcRn produced by GeneArt AG (Germany) (diluted to 10 ฮผg/mL in 10 mM sodium acetate pH5.0 (G E Healthcare)) was immobilised on flow cell 2 (FC2) to levels between 1600-2200 response units (RU) via standard amine coupling as per manufacturers instructions (G E Healthcare). A blank immobilisation was performed on flow cell 1 (FC1) for it to serve as a reference cell. To stabilise the assay, 3-5 start up cycles were run first, with running buffer (67 mM phosphate buffer, 0.15M NaCl, 0.005% Tween 20 at pH5.75ยฑ0.25) only, followed by regeneration. WT rHA and K500 library variants were injected at various concentrations (1 ฮผM-150 ฮผM) for 90 s at a constant flow rate of (30 ฮผl/min) at 25ยฐ C. followed by regeneration of the surface using HBS-EP buffer pH7.4 (G E Healthcare) until approximate initial baseline RU was restored (usually 12 s pulse would suffice).
  • Results are shown in the Table 17 and FIG. 16
  • TABLE 17
    Kinetics of HSA K500 single point mutants.
    KD
    Albumin ka kd KDb Reqc
    variant (103/Ms) (10โˆ’3/s) (ฮผM) (ฮผM)
    K500R 4.42 7.21 1.63
    K500I 5.18 10.9 2.1
    WT 4.24 9.2 2.2a
    K500L 3.73 11.9 3.2
    K500Q 1.07 3.4 3.2
    K500V 3.29 11.0 3.3
    K500Y 3.97 14.6 3.7
    K500M 2.48 21.5 8.7
    K500T 1.2 13.4 11.2
    K500W 0.5 5.4 11.7
    K500N 1.3 18.2 14
    K500F 5.17 73.7 14.3
    K500H 4 63.8 16
    K500P ND ND ND 51*
    K500C 2.38 124 52
    K500S ND ND โˆ’ ND 70.2*
    K500A 2.61 208 79.9
    K500D ND ND ND 83.3*
    K500G ND ND ND 95.4*
    K500E KD not calculable see FIG. 16
    K500 STOP Null binder
    aMean of 4 values.
    bThe kinetic rate constants were obtained using a simple first-order (1:1) bimolecular interaction model.
    cThe steady state affinity constant was obtained using an equilibrium (Req) binding model supplied by the BIAevaluation 4.1 software.
  • The results show for variants K500R and K500I have increased and comparable affinity for shFcRn compared to WT HSA respectively. Variant K500E bound tightly to immobilised shFcRn but still demonstrated the characteristic pH-dependency of the FcRn interaction. This complex was very stable, such that kinetic analysis was not possible (FIG. 16). All other variants have reduced binding to shFcRn than wt rHA.
  • All variants bound to shFcRn (to some extent) at pH5.5. No binding of K500 library variants to shFcRn was detectable at pH7.4.
  • Example 16. Fusion Polypeptides
  • The Generation of Albumin Fusions Containing Albumin Muteins
  • Plasmids containing expression cassettes for the production of scFv (vHvL) genetically-fused to HSA, at either the N- or C-terminus or both, (described in, Evans et al., 2010. Protein Expression and Purification. 73, 113-124) were modified to allow the production of albumin fusions using in vivo cloning (describe above). That is, pDB3017 (FIG. 17), pDB3021 (FIG. 18), pDB3056 (FIG. 19) were digested with NsiI/SpeI and NsiI fragments corresponding 9.511 kb, 9.569 kb and 8.795 kb, respectively, were purified using standard techniques. Purified NsiI fragments were self-ligated and used to transform chemically competent E. coli DH5ฮฑ to produce pDB4168, pDB4169 and pDB4170, respectively (Table 18).
  • Similarly, pDB3165 (containing the bivalent fusion) (FIG. 20) was digested with NotI and the expression cassette (4.506 kb fragment) was purified before being ligated into NotI-digested pDB3927 to produce pDB4172 (FIG. 21, Table 18).
  • Synthetic SalI/Bsu36I DNA fragments (269 bp), which contain point mutations within the albumin encoding nucleotide sequence to introduce amino acid substitutions corresponding to K500A, or D550N or K573P into the translated albumin protein sequence, were generated by gene assembly (GeneArt AG, Germany). The SalI/Bsu36I fragments were individually ligated into SalI/Bsu36I-digested pDB4168-pDB4170 and pDB4172 and used to transform chemically competent E. coli DH5ฮฑ using standard techniques to generate plasmids pDB4265-pDB4276 (Table 18).
  • TABLE 18
    Albumin variant fusions
    Plasmid Construct
    pDB3017 scFv (anti-FITC)-HSA-FLAG
    pDB3021 HSA-GS linker-scFv (anti-FITC)-FLAG
    pDB3056 HSA-FLAG
    pDB3165 scFv (anti-FITC)-HSA-GS linker-scFv (anti-FITC)-FLAG
    pDB4168 scFv (anti-FITC)-HSA-FLAG
    pDB4169 HSA-GS linker-scFv (anti-FITC)-FLAG
    pDB4170 HSA-FLAG
    pDB4172 scFv (anti-FITC)-HSA-GS linker-scFv (anti-FITC)-FLAG
    pDB4265 scFv (anti-FITC)-HSA K500A-FLAG
    pDB4266 scFv (anti-FITC)-HSA D550N-FLAG
    pDB4267 scFv (anti-FITC)-HSA K573P-FLAG
    pDB4268 HSA K500A-GS linker-scFv (anti-FITC)-FLAG
    pDB4269 HSA D550N-GS linker-scFv (anti-FITC)-FLAG
    pDB4270 HSA K573P-GS linker-scFv (anti-FITC)-FLAG
    pDB4271 HSA K500A-FLAG
    pDB4272 HSA D550N-FLAG
    pDB4273 HSA K573P-FLAG
    pDB4274 scFv (anti-FITC)-HSA K500A-GS linker-scFv (anti-FITC)-
    FLAG
    pDB4275 scFv (anti-FITC)-HSA D550N-GS linker-scFv (anti-FITC)-
    FLAG
    pDB4276 scFv (anti-FITC)-HSA K573P-GS linker-scFv (anti-FITC)-
    FLAG
    pDB4277 scFv (anti-FITC)-HSA K573A-FLAG
    pDB4278 HSA K573A-GS linker-scFv (anti-FITC)-FLAG
    pDB4279 HSA K573A-FLAG
    pDB4280 scFv (anti-FITC)-HSA K573A-GS linker-scFv (anti-FITC)-
    FLAG
    pDB4281 HSA K500A-GS linker-scFv (anti-FITC)
    pDB4282 HSA D550N-GS linker-scFv (anti-FITC)
    pDB4283 HSA K573P-GS linker-scFv (anti-FITC)
    pDB4284 HSA-GS linker-scFv (anti-FITC)
    pDB2613 HSA-GS linker-IL1RA (N84Q)
    pDB4285 HSA K573A-GS linker-IL1RA (N84Q)
    pDB4286 HSA D550N-GS linker-IL1RA (N84Q)
    pDB4287 HSA K500A-GS linker-IL1RA (N84Q)
    pDB4288 HSA K573P-S linker-IL1RA (N84Q)
  • Similarly, a DNA fragment was generated by PCR (using standard techniques), to introduce a K573A substitution in the translated albumin protein sequence. PCR was performed using the New England Biolabs Phusion kit using pDB4267 (FIG. 22) as template DNA and oligonucleotides xAP238 (SEQ ID NO: 53) and xAP239 (SEQ ID NO: 54):
  • Table 19 describes PCR cycling.
  • TABLE 19
    PCR cycling
    98ยฐ C. for 2 min โ€‚1 cycle
    98ยฐ C. for 10 sec 35 cycles
    57ยฐ C. for 30 sec
    72ยฐ C. for 10 sec
    72ยฐ C. for 5 min โ€‚1 cycle
  • The PCR-product was purified, digested with SalI/Bsu36I, and the fragment (269 bp) isolated was ligated into SalI/Bsu36I-digested pDB4168-pDB4170 and pDB4172 and used to transform chemically competent E. coli DH5ฮฑ. Resulting plasmids (pDB4277-pDB4280) are listed in Table 18.
  • The nucleotide sequence encoding the FLAG tag was removed from plasmids pDB4168 and pDB4268-4270 (plasmids for the expression of scFv N-terminally fused to HSA and HSA muteins K500A, D550N and K573P, respectively. pDB4168 and pDB4268-4270 (Table 18) were digested with Bsu36I/SphI to remove a 231 bp product comprising 3โ€ฒ region of HSA-encoding gene, nucleotide sequence encoding FLAG tag and 5โ€ฒ region of ADH1 terminator. A Bsu36I/SphI fragment (207 bp), comprising 3โ€ฒ region of HSA-encoding gene and 5โ€ฒ region of mADH1 terminator (SEQ ID1) from pDB4181 was ligated into Bsu36I/SphI-digested pDB4168 and pDB4268-pDB4270 using standard techniques. Ligation mixtures were used to transform chemically competent E. coli DH5ฮฑ using standard techniques to generate plasmids pDB4281-pDB4284 (Table 18)
  • pDB4265-pDB4284 were digested with BstEII/BsrBI and the linearised DNA molecules were purified using standard techniques. One hundred ng BstEII/BsrBI DNA samples were mixed with 100 ng Acc65I/BamHI-digested pDB3936 and used to transform S. cerevisiae BXP10cir0 using the Sigma Yeast Transformation kit described below. In each case the expression plasmid was generated in the yeast by homologous recombination (in vivo cloning) between the albumin-fusion containing plasmid (pDB4265-pDB4280) (Table 18) and pDB3936.
  • Plasmids pDB3017, pDB3021, pDB3056 and pDB3165 (wild type HSA fusions, described by Evans et al., 2010. Protein Expression and Purification. 73, 113-124) were used to transform S. cerevisiae Strain Acir0 (described in WO/2005/061718) using the Sigma Yeast Transformation kit described below.
  • The nucleotide sequence encoding human IL-1RA (interleukin-1 receptor antagonist) (accession number: CAA59087) could be synthetically generated by gene assembly. The nucleotide sequence of the 708 bp synthetic fragment (Bsu36I/SphI fragment) is given in SEQ ID NO: 55 and includes the 3โ€ฒ region of the gene encoding HSA, the nucleotide sequence encoding a GS linker, the nucleotide sequence encoding human IL-1RA (N84Q to abolish the N-linked glycosylation motif) and the 5โ€ฒ region of the ADH1 terminator. The synthetic DNA fragment could be ligated into Bsu36I/SphI-digested pDB3927 to produce pDB2588.
  • Plasmids containing the expression cassettes for the production of IL-1RA genetically fused to the C-terminus of HSA and the HSA variants K500A, D550N, K573A and K573P were prepared as follows. pDB2588 was digested with Bsu36I/SphI and a 705 bp fragment containing the โ€˜3 region of the HSA encoding gene, nucleotide sequence encoding a GS linker, nucleotide sequence encoding human IL1-RA (N84Q) and the 5โ€™ region of a modified S. cerevisiae ADH1 terminator (SEQ 1D3) was purified using standard techniques then ligated into Bsu36I/SphI-digested pDB4006 (containing HSA K573A expression cassette), pDB4010 (containing HSA D550N expression cassette), pDB4086 (containing HSA K500A expression cassette), pDB4110 (containing HSA K573P expression cassette) to generate pDB4287, pDB4286, pDB4285 and pDB4288, respectively (for an example, see FIG. 23). pDB4285-pDB4288 were digested with NsiI/PvuI and the linearised DNA molecules were purified using standard techniques. One hundred ng NsiI/PvuI-digested DNA samples were mixed with 100 ng Acc65I/BamHI-digested pDB3936 (9721 bp) (i.e. in vivo cloning) and used to transform S. cerevisiae (i.e. by in vivo cloning) using the Sigma Yeast Transformation kit described below.
  • Preparation of an S. cerevisiae strain expressing wild type HSA genetically fused to a GS linker and IL1-RA (N84Q) (see Table 18) could also be generated following the methods described above.
  • The fusion polypeptides were analysed for their binding to FcRn using the SPR method described above and following results were obtained:
  • TABLE 20
    Kinetics of HSA fusion variants.
    ka kd KDb
    Albumin varianta (103/Ms) (10โˆ’3/s) (ฮผM)
    HSAWT 9.7 ยฑ 0.0 30.0 ยฑ 0.1โ€‚ 3.1
    K574N 4.9 ยฑ 01โ€‰ 8.4 ยฑ 0.1 1.7
    Q580K 6.0 ยฑ 0.0 9.3 ยฑ 0.0 1.5
    K573P 2.8 ยฑ 0.0 0.4 ยฑ 0.0 0.1
    HSA-WT-FLAG 8.2 ยฑ 0.2 24.0 ยฑ 0.2โ€‚ 2.9
    HSA-D550N-FLAG 5.9 ยฑ 0.0 49.0 ยฑ 0.1โ€‚ 8.3
    HSA-K500A-FLAG NDc ND ND
    HSA-K573A-FLAG 6.1 ยฑ 0.1 7.1 ยฑ 0.1 1.1
    HSA-K573P-FLAG 6.2 ยฑ 0.1 1.2 ยฑ 0.1 0.2
    HSA-WT-IL1RA 6.2 ยฑ 0.0 25.0 ยฑ 0.2โ€‚ 4.0
    HSA-K500A-IL1RA ND ND ND
    HSA-D550N-IL1RA 7.3 ยฑ 0.2 38.0 ยฑ 0.0โ€‚ 5.2
    HSA-K573A-IL1RA 6.1 ยฑ 0.0 7.1 ยฑ 0.1 1.1
    HSA-K573P-IL1RA 6.2 ยฑ 0.1 1.3 ยฑ 0.1 0.2
    scFv-HSA-K500A-FLAG ND ND ND
    scFv-HSA-D550N-FLAG 6.2 ยฑ 0.0 18.0 ยฑ 0.0โ€‚ 2.9
    scFv-HSA-K573A-FLAG 6.4 ยฑ 0.1 5.7 ยฑ 0.2 0.9
    scFv-HSA-K573P-FLAG 5.8 ยฑ 0.0 1.1 ยฑ 0.1 0.2
    scFv-HSA-WT-scFv- 7.5 ยฑ 0.0 15.0 ยฑ 0.2โ€‚ 2.0
    FLAG
    scFv-HSA-K500A-scFv- ND ND ND
    FLAG
    scFv-HSA-D550N-scFv- 4.1 ยฑ 0.1 27.0 ยฑ 0.2โ€‚ 6.6
    FLAG
    scFv-HSA-K573P-scFv- 6.0 ยฑ 0.2 0.7 ยฑ 0.1 0.1
    FLAG
    HSA-K500A-scFv-FLAG ND ND ND
    HSA-D550N-scFv-FLAG 7.3 ยฑ 0.1 42.0 ยฑ 0.3โ€‚ 5.8
    HSA-K573A-scFv-FLAG 6.4 ยฑ 0.1 5.7 ยฑ 0.1 0.9
    HSA-K573P-scFv-FLAG 4.7 ยฑ 0.1 0.7 ยฑ 0.1 0.1
    scFv-HSA-K500A ND ND ND
    scFv-HSA-D550N 7.5 ยฑ 0.1 19.0 ยฑ 0.2โ€‚ 2.5
    scFv-HSA-K573P 7.4 ยฑ 0.1 0.8 ยฑ 0.1 0.1
    aDilutions of HSA variants were injected over immobilized shFcRn (~1500 RU).
    bThe kinetic rate constants were obtained using a simple first-order (1:1) bimolecular interaction model. The kinetic values represent the average of duplicates.
    cNot determined due to weak binding (ND).
  • In example 8 it was shown that the K500A variant did not significantly bind shFcRn, in Example 10 it was shown that the K573P and K573A variants bind shFcRn stronger than HSA and in Example 11 it was shown that the D550N variant binds FcRn weaker than HSA.
  • In the present example it is shown that these observed difference in binding properties also are reflected in fusion polypeptides in different configurations: C-terminal fusions with a small moiety (HSA-FLAG), C-terminal fusions with a larger polypeptide (HSA-IL1RA); N-terminal fusions with polypeptide (scFv-HSA); N- and C-terminal fusions (scFv-HSA-FLAG and scFv-HSA-scFv-FLAG).
  • Example 17. Conjugation of Horseradish Peroxidase Protein to Albumin and the K573P Variant
  • For conjugation analysis, commercially available recombinant albumin (Recombuminโ„ข) was used as a control molecule. For this example, a final 200 mg/mL albumin K573P variant of the invention was purified from a fed batch fermentation by means described in Material and Methods. A two step purification was carried out;
  • The first step used a column (bed volume approximately 400 mL, bed height 11 cm) packed with AlbuPureโ„ข matrix (ProMetic). This was equilibrated with 50 mM sodium acetate, pH 5.3 and loaded with neat culture supernatant, at approximately pH 5.5-6.5, to approximately 20 mg/mL matrix. The column was then washed with approximately 5 column volumes each of 50 mM sodium acetate, pH 5.3, 50 mM sodium phosphate, pH 6.0, 50 mM sodium phosphate, pH 7.0 and 50 mM ammonium acetate, pH 8.0, respectively. Bound protein was eluted using approximately two column volumes of 50 mM ammonium acetate, 10 mM octanoate, pH 7.0. The flow rate for the entire purification was 154 mL/min.
  • For the second step, the eluate from the first step was diluted approximately two fold with water to give a conductivity of 2.5ยฑ0.5 mS/cm after adjustment to pH 5.5ยฑ0.3 with acetic acid. This was loaded onto a DEAE-Sepharose Fast Flow (GE Healthcare) column (bed volume approximately 400 mL, bed height 11 cm), equilibrated with 80 mM sodium acetate, 5 mM octanoate, pH 5.5. Loading was approximately 30 mg protein/mL matrix. The column was washed with approximately 5 column volumes of 80 mM sodium acetate, 5 mM octanoate, pH 5.5. Followed by approximately 10 column volumes of 15.7 mM potassium tetraborate, pH 9.2. The bound protein was eluted using two column volumes of 110 mM potassium tetraborate, 200 mM sodium chloride, approximately pH 9.0. The flow rate was 183 mL/min during the load and wash steps, and 169 mL/min during the elution step.
  • The eluate was concentrated and diafiltered against 145 mM NaCl, using a Pall Centramate Omega 10,000 Nominal MWCO membrane, to give a final protein concentration of approximately 200 mg/mL.
  • Both 200 mg/mL stock solutions of the rHA and K573P variant albumin were diluted down to 5 mg/mL, using phosphate buffer saline (PBS), pH adjusted to pH 6.5-6.7. This ensured a favourable pH environment for the maleimide reactive group of the EZ-Linkยฎ Maleimide Activated Horseradish Peroxidase (Thermo Scientific) to react with the free sulphydryl, to form a stable thioester bond. 2 mg of the EZ-Linkยฎ Maleimide Activated Horseradish Peroxidase (HRP) was mixed with either 1 mL of the 5 mg/ML rHA or K573P variant albumin. This mixture ensured an approximate 2 fold molar excess of the albumin, or K573P variant albumin. This mixture was minimally incubated at 4ยฐ C., for 24 hours. The reaction mixtures were then checked for conjugation, using GP-HPLC.
  • To separate unconjugated species (rHA, or Albumin variant K573P and unreacted HRP) from the corresponding conjugated species the samples were first concentrated (Vivaspin20, 10,000 MWCO PES, Sartorius), and then individually applied to a Tricorn Superdexโ„ข 200, 10/300 GL column (GE Healthcare), run at a flow rate of 45 cm/hr in PBS. The elution peak was fractionated and GP-HPLC analysed. Fractions containing the conjugated species were pooled, concentrated and diafiltered against 50 mM NaCl and analysed by GP-HPLC to demonstrate (FIG. 24) These samples were then assayed using the Biacore method described herein (Table 21).
  • This example demonstrates that the K573P maintains its increased affinity for shFcRn compared the the WT HSA.
  • Example 18. Conjugation of Fluorescein to Albumin and the K573P Variant
  • The two same albumin samples used in Example 17, were also the start materials for this example. I.e. Approximately 200 mg/mL rHA or the K573P albumin variant.
  • Fluorescein-5-Maleimide, Thermo Scientific (F5M) was dissolved in dimethylformamide, to give a final concentration of 25 mg/mL. This was then further diluted into 18 mls of PBS, pH adjusted to approximately pH 6.5. To this solution either 1 ml of 200 mg/mL rHA or 1 mL of 200 mg/mL K573P variant was added. This gave an approximate 20 fold final molar excess of F5M. These samples were incubated and allowed to conjugate overnight at 4ยฐ C., in the dark, to allow the maleimide groups on the F5M to react with predominantly the free sulfhydryl, present in both albumin species.
  • Following overnight incubation aliquots of the reaction mixtures were extensively diafiltered against 50 mM NaCl to remove unconjugated F5M, (Vivaspin20, 10,000 MWCO PES, Sartorius). Conjugation was confirmed by ultraviolet visualization of conjugated Fluorescein::Albumins Following standard SDS-PAGE (FIG. 25).
  • These diafiltered samples were then assayed using the Biacore method described herein (Table 21). This example demonstrates that the conjugation of a small molecule to either rHA or a variant, e.g. K573P does not affect the trend in binding affinities to shFcRn.
  • TABLE 21
    Representative Biacore assay KD values of
    conjugated rHA or a variant (K573P) when
    binding to immobilized shFcRn.
    Analyte KD (ฮผM)
    rHA::HRP 3.6
    K573P::HRP 0.02
    rHA::F5M 7.3
    K573P::F5M 2.5
  • Example 19. Further Albumin Variants
  • The following variants were generated using the methods described in Example 1 E492T, N503D, E492T+N503D, K538H, E542D, D494N+E495Q+T496A, E495Q+T496A, N403K, K541A and K541N. SPR analysis was carried out as described in Example 15 and the results presented in FIG. 26 and FIG. 27.
  • FIGS. 30A and 30B shows the effect on shFcRn binding for the albumin variants.
    Substitutions N503D, D494N+E495Q+T496A E492T+N503D, E495Q+T496A within HSA had a negative impact on binding to shFcRn at pH5.5.
  • Example 20. Variants of Albumin at the C-Termini
  • The following variants were generated using the methods described in Example 1. Binding to the shFcRn was determined as described in Materials and Methods and the results are presented in Table 22.
  • TABLE 22
    Kinetics of the HSA C-terminal swapped variant
    interactions with shFcRn.
    Albumin ka kd KDb
    varianta (103/Ms) (10โˆ’3/s) (ฮผM)
    HSA 4.4 ยฑ 0.0 24.0 ยฑ 0.1โ€‚ 5.4
    MacSA 3.1 ยฑ 0.1 8.6 ยฑ 0.1 2.7
    HSA-MacC 4.1 ยฑ 0.1 5.6 ยฑ 0.0 1.3
    MouseSAc 3.8 ยฑ 0.0 3.1 ยฑ 0.1 0.8
    HSA-MouseC 3.7 ยฑ 0.1 1.3 ยฑ 0.0 0.3
    RabbitSAd 1.9 ยฑ 0.3 1.7 ยฑ 0.1 0.9
    HSA-RabC 3.5 ยฑ 0.0 1.6 ยฑ 0.0 0.4
    SheepSA ND ND ND
    HSA-SheepC 3.3 ยฑ 0.0 2.1 ยฑ 0.0 0.6
    aDilutions of HSA variants were injected over immobilized shFcRn (~1500 RU).
    bThe kinetic rate constants were obtained using a simple first-order (1:1) bimolecular interaction model.
    cData from Table 2
    dData from Table 3
    Not determined due to weak binding (ND)
  • This example demonstrates that for all C-terminal swaps to human albumin tested an increase in binding over the donor albumin was observed. All donor sequences contain the K573P substitution shown to significantly increase binding but less that the K573P alone (Table 20).
  • Example 21. Competitive Binding Analysis of Variant Albumin Fusions
  • Competitive binding studies, using variant albumin fusions and a selection of variant albumins prepared as described in Example 1, were performed as described in Example 4. Results are presented in FIGS. 28-31.
  • The competitive binding hierarchy was identical for the variants fusions of HSA-FLAG and, N+C-terminal scFv HSA-FLAG to the hierarchy of the individual HSA variants (unfused and fused) affinity data. For the IL1 Ra variants K573P, K573A, and the K500A were as predicted, however the D550N appears to inhibit more efficiently than the WT fusion.
  • Example 22. Further HSA Variants
  • The following variants were generated using methods described in Example 1: HSA E492G+K573A, HSA E492G+N503K+K573A, HSA E492G+N503H+K573A, HSA E492G+K573P, HSA E492G+N503K+K573P, HSA E492G+N503H+K573P. SPR analysis was performed as described in Materials and Methods. Results (FIG. 32) showed that all HSA variants bound more strongly to shFcRn compared to wild type HSA at pH 5.5. No binding was observed at pH 7.4.
  • HSA E492G+K573A, HSA E492G+N503K+K573A, unlike HSA E492G+N503H+K573A, had marginally improved binding beyond that of HSA K573A. The combination variants containing K573P did not show improved binding over the K573P single variant.

Claims (1)

1. A Method for preparing a variant of albumin, fragments thereof or fusion polypeptide comprising said variant albumin or fragment thereof, comprising following steps:
a. Providing a nucleic acid encoding a parent albumin having at least 80% sequence identity to SEQ ID NO: 2;
b. Modifying the sequence of step a., to encode a variant albumin, fragments thereof or fusion polypeptide comprising said variant albumin or fragment thereof having one or more substitutions corresponding to the substitutions in SEQ ID NO: 2 selected among:
Q417A,C,D,E,F,G,H,I,K,L,M,N,P,R,S,T,V,W,Y;
H440A,C,D,E,F,G,I,K,L,M,N,P,Q,R,S,T,V,W,Y;
A490C,D,E,F,G,H,I,K,L,M,N,P,R,S,T,V,W,Y
E492A,C,D,F,G,H,I,K,L,M,N,P,Q,R,S,T,V,W,Y;
V493A,C,D,E,F,G,H,I,K,L,M,N,P,Q,R,S,T.W,Y;
D494A,C,E,F,G,H,I,K,L,M,N,P,Q,R,S,T,V,W,Y;
E495A,C,D,F,G,H,I,K,L,M,N,P,Q,R,S,T,V,W,Y;
T496A,C,D,E,F,G,H,I,K,L,M,N,P,Q,R,S,V,W,Y;
P499A,C,D,E,F,G,H,I,K,L,M,N,Q,R,S,T,V,W,Y;
K500A,C,D,E,F,G,H,I,L,M,N,Q,R,S,T,V,W,Y;
E501A,C,D,F,G,H,I,K,L,M,N,P,Q,R,S,T,V,W,Y;
N503A,C,D,E,F,G,H,I,K,L,M,P,Q,R,S,T,V,W,Y;
A504C,D,F,G,H,I,K,L,M,N,P,Q,R,S,T,V,W,Y;
E505A,C,D,F,G,H,I,K,L,M,N,P,Q,R,S,T,V,W,Y;
T506A,C,D,F,G,H,I,K,L,M,N,P,Q,R,S,V,W,Y;
H510A,C,D,F,G,I,K,L,M,N,P,Q,R,S,T,V,W,Y;
H535A,C,D,F,G,I,K,L,M,N,P,Q,R,S,T,V,W,Y;
K536A,C,D,E,F,G,H,I,L,M,N,P,Q,R,S,T,V,W,Y;
P537A,C,D,E,F,G,H,I,K,L,M,N,Q,R,S,T,V,W,Y;
K538A,C,D,E,F,G,H,I,L,M,N,P,Q,R,S,T,V,W,Y;
T540A,C,D,E,F,G,H,I,L,M,N,P,Q,R,S,V,W,Y;
K541A,C,D,E,F,G,H,I,L,M,N,P,Q,R,S,T,V,W,Y;
E542A,C,D,F,G,H,I,K,L,M,N,P,Q,R,S,T,V,W,Y;
D550A,C,E,F,G,H,I,K,L,M,N,P,Q,R,S,T,V,W,Y;
K573A,C,D,E,F,G,H,I,L,M,N,P,Q,R,S,T,V,W,Y;
K574A,C,D,E,F,G,H,I,L,M,N,P,Q,R,S,T,V,W,Y;
Q580A,C,D,E,F,G,H,I,K,L,M,N,P,R,S,T,V,W,Y;
A581C,D,E,F,G,H,I,K,L,M,N,P,Q,R,S,T,V,W,Y;
A582C,D,E,F,G,H,I,K,L,M,N,P,Q,R,S,T,V,W,Y; or
G584A,C,D,E,F,H,I,K,L,M,N,P,Q,R,S,T,V,W,Y;
c. Introducing the modified sequence of step b., in a suitable host cell;
d. Growing the cells in a suitable growth medium under condition leading to expression of the variant of albumin, fragments thereof or fusion polypeptide comprising said variant albumin or fragment thereof; and
e. Recovering the variant of albumin, fragments thereof or fusion polypeptide comprising said variant albumin or fragment thereof from the growth medium;
wherein the variant of albumin, fragments thereof or fusion polypeptide comprising said variant albumin or fragment thereof, has an altered plasma half-life compared with the parent albumin, fragments thereof or fusion polypeptide comprising said variant albumin or fragment thereof.
US15/979,319 2009-10-30 2018-05-14 Albumin variants Active US10696732B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US15/979,319 US10696732B2 (en) 2009-10-30 2018-05-14 Albumin variants
US16/882,019 US20200385442A1 (en) 2009-10-30 2020-05-22 Albumin variants

Applications Claiming Priority (13)

Application Number Priority Date Filing Date Title
EP09174698 2009-10-30
EP09174698 2009-10-30
EP09174698.2 2009-10-30
US32717110P 2010-04-23 2010-04-23
US34800110P 2010-05-25 2010-05-25
EP10174162 2010-08-26
EP10174162.7 2010-08-26
EP10174162 2010-08-26
PCT/EP2010/066572 WO2011051489A2 (en) 2009-10-30 2010-11-01 Albumin variants
US201213504326A 2012-04-26 2012-04-26
US14/262,244 US20140234311A1 (en) 2009-10-30 2014-04-25 Albumin variants
US14/863,719 US20160075759A1 (en) 2009-10-30 2015-09-24 Albumin variants
US15/979,319 US10696732B2 (en) 2009-10-30 2018-05-14 Albumin variants

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US14/863,719 Division US20160075759A1 (en) 2009-10-30 2015-09-24 Albumin variants

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/882,019 Division US20200385442A1 (en) 2009-10-30 2020-05-22 Albumin variants

Publications (2)

Publication Number Publication Date
US20180265570A1 true US20180265570A1 (en) 2018-09-20
US10696732B2 US10696732B2 (en) 2020-06-30

Family

ID=43922679

Family Applications (19)

Application Number Title Priority Date Filing Date
US13/504,326 Active US8748380B2 (en) 2009-10-30 2010-11-01 Albumin variants
US14/262,244 Abandoned US20140234311A1 (en) 2009-10-30 2014-04-25 Albumin variants
US14/863,752 Abandoned US20160075760A1 (en) 2009-10-30 2015-09-24 Albumin variants
US14/863,628 Abandoned US20160009787A1 (en) 2009-10-30 2015-09-24 Albumin variants
US14/863,655 Abandoned US20160075757A1 (en) 2009-10-30 2015-09-24 Albumin variants
US14/863,719 Abandoned US20160075759A1 (en) 2009-10-30 2015-09-24 Albumin variants
US14/863,685 Abandoned US20160075758A1 (en) 2009-10-30 2015-09-24 Albumin variants
US14/863,597 Abandoned US20160075756A1 (en) 2009-10-30 2015-09-24 Albumin variants
US14/863,780 Abandoned US20160075761A1 (en) 2009-10-30 2015-09-24 Albumin variants
US14/863,836 Abandoned US20160075762A1 (en) 2009-10-30 2015-09-24 Albumin variants
US14/863,868 Abandoned US20160075763A1 (en) 2009-10-30 2015-09-24 Albumin variants
US15/709,263 Abandoned US20180105578A1 (en) 2009-10-30 2017-09-19 Albumin variants
US15/709,169 Abandoned US20180105576A1 (en) 2009-10-30 2017-09-19 Albumin variants
US15/709,227 Abandoned US20180072792A1 (en) 2009-10-30 2017-09-19 Albumin variants
US15/709,261 Abandoned US20180105577A1 (en) 2009-10-30 2017-09-19 Albumin variants
US15/870,376 Abandoned US20180222963A1 (en) 2009-10-30 2018-01-12 Albumin variants
US15/870,349 Abandoned US20180162925A1 (en) 2009-10-30 2018-01-12 Albumin variants
US15/979,319 Active US10696732B2 (en) 2009-10-30 2018-05-14 Albumin variants
US16/882,019 Abandoned US20200385442A1 (en) 2009-10-30 2020-05-22 Albumin variants

Family Applications Before (17)

Application Number Title Priority Date Filing Date
US13/504,326 Active US8748380B2 (en) 2009-10-30 2010-11-01 Albumin variants
US14/262,244 Abandoned US20140234311A1 (en) 2009-10-30 2014-04-25 Albumin variants
US14/863,752 Abandoned US20160075760A1 (en) 2009-10-30 2015-09-24 Albumin variants
US14/863,628 Abandoned US20160009787A1 (en) 2009-10-30 2015-09-24 Albumin variants
US14/863,655 Abandoned US20160075757A1 (en) 2009-10-30 2015-09-24 Albumin variants
US14/863,719 Abandoned US20160075759A1 (en) 2009-10-30 2015-09-24 Albumin variants
US14/863,685 Abandoned US20160075758A1 (en) 2009-10-30 2015-09-24 Albumin variants
US14/863,597 Abandoned US20160075756A1 (en) 2009-10-30 2015-09-24 Albumin variants
US14/863,780 Abandoned US20160075761A1 (en) 2009-10-30 2015-09-24 Albumin variants
US14/863,836 Abandoned US20160075762A1 (en) 2009-10-30 2015-09-24 Albumin variants
US14/863,868 Abandoned US20160075763A1 (en) 2009-10-30 2015-09-24 Albumin variants
US15/709,263 Abandoned US20180105578A1 (en) 2009-10-30 2017-09-19 Albumin variants
US15/709,169 Abandoned US20180105576A1 (en) 2009-10-30 2017-09-19 Albumin variants
US15/709,227 Abandoned US20180072792A1 (en) 2009-10-30 2017-09-19 Albumin variants
US15/709,261 Abandoned US20180105577A1 (en) 2009-10-30 2017-09-19 Albumin variants
US15/870,376 Abandoned US20180222963A1 (en) 2009-10-30 2018-01-12 Albumin variants
US15/870,349 Abandoned US20180162925A1 (en) 2009-10-30 2018-01-12 Albumin variants

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/882,019 Abandoned US20200385442A1 (en) 2009-10-30 2020-05-22 Albumin variants

Country Status (14)

Country Link
US (19) US8748380B2 (en)
EP (2) EP3421491A3 (en)
JP (3) JP2013509170A (en)
KR (1) KR101874834B1 (en)
CN (2) CN102741280B (en)
AU (1) AU2010311332B2 (en)
BR (1) BR112012009450A2 (en)
CA (1) CA2776241A1 (en)
ES (1) ES2700230T3 (en)
GB (1) GB2488077A (en)
IL (1) IL218858B (en)
MX (1) MX2012004793A (en)
RU (1) RU2607374C2 (en)
WO (1) WO2011051489A2 (en)

Cited By (5)

* Cited by examiner, โ€  Cited by third party
Publication number Priority date Publication date Assignee Title
US10329340B2 (en) 2012-03-16 2019-06-25 Albumedix Ltd Albumin variants
US10501524B2 (en) 2012-11-08 2019-12-10 Albumedix Ltd Albumin variants
US10633428B2 (en) 2015-08-20 2020-04-28 Albumedix Ltd Albumin variants and conjugates
US10711050B2 (en) 2011-11-18 2020-07-14 Albumedix Ltd Variant serum albumin with improved half-life and other properties
US11555061B2 (en) 2009-02-11 2023-01-17 Albumedix, Ltd Albumin variants and conjugates

Families Citing this family (108)

* Cited by examiner, โ€  Cited by third party
Publication number Priority date Publication date Assignee Title
CA2417960C (en) 2000-08-04 2012-07-10 Dmi Biosciences, Inc. Method of using diketopiperazines and composition containing them
CN104095851A (en) 2003-05-15 2014-10-15 ๅฎ‰็šฎๅฅฅๅˆถ่ฏ่‚กไปฝๆœ‰้™ๅ…ฌๅธ Treatment of T-cell mediated diseases
US8217047B2 (en) 2008-05-27 2012-07-10 Dmi Acquisition Corp. Therapeutic methods and compounds
BR112012009450A2 (en) 2009-10-30 2017-05-23 Novozymes Biopharma Dk As albumin variants
EP2536756B1 (en) * 2010-02-16 2018-04-25 MedImmune, LLC Hsa-related compositions and methods of use
CN102939304B (en) 2010-04-09 2017-04-19 ้˜ฟๅฐ”ๅธƒ้บฆ็‹„ๅ…‹ๆ–ฏๅ…ฌๅธ albumin derivatives and variants
WO2011146902A1 (en) 2010-05-21 2011-11-24 Merrimack Pharmaceuticals, Inc. Bi-specific fusion proteins
MY192639A (en) 2010-07-09 2022-08-29 Bioverativ Therapeutics Inc Factor ix polypeptides and methods of use thereof
US20130225496A1 (en) * 2010-11-01 2013-08-29 Novozymes Biopharma Dk A/S Albumin Variants
US9045564B2 (en) 2011-02-15 2015-06-02 Medimmune, Llc HSA-related compositions and methods of use
WO2012116453A1 (en) 2011-03-03 2012-09-07 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs
EP2705051A1 (en) 2011-05-05 2014-03-12 Novozymes Biopharma DK A/S Albumin variants
EP2729492B1 (en) 2011-07-05 2019-01-02 Albumedix Ltd Albumin formulation and use
AU2012286048A1 (en) 2011-07-18 2014-02-20 Arts Biologics A/S Long acting luteinizing hormone (LH) compound
CN103841987B (en) 2011-10-10 2015-06-17 ๅฎ‰็šฎๅฅฅๅˆถ่ฏ่‚กไปฝๆœ‰้™ๅ…ฌๅธ Treatment of degenerative joint disease
BR112014007657A2 (en) 2011-10-10 2017-04-11 Ampio Pharmaceuticals Inc Implantable medical devices with improved immune tolerance and methods for production and implantation
KR102132997B1 (en) 2011-10-11 2020-07-14 ๋ฉ”๋””๋ฎจ ์—˜์—˜์”จ Cd40l-specific tn3-derived scaffolds and methods of use thereof
BR112014008036A2 (en) 2011-10-28 2017-04-11 Ampio Pharmaceuticals Inc rhinitis treatment
EP2846822A2 (en) 2012-05-11 2015-03-18 Prorec Bio AB Method for diagnosis and treatment of prolactin associated disorders
WO2014005596A1 (en) 2012-07-03 2014-01-09 Aarhus Universitet Modified payload molecules and their interactions and uses
AU2013289881B2 (en) 2012-07-13 2018-01-18 Zymeworks Bc Inc. Multivalent heteromultimer scaffold design and constructs
SG11201505715RA (en) * 2013-02-01 2015-08-28 Ampio Pharmaceuticals Inc Methods for producing diketopiperazines and compositions containing diketopiperazines
KR20150118123A (en) 2013-02-16 2015-10-21 ๋…ธ๋ณด์ž์ž„์Šค ๋ฐ”์ด์˜คํŒŒ๋งˆ ๋””์ผ€์ด ์—์ด/์—์Šค Pharmacokinetic animal model
AU2014228938B2 (en) 2013-03-15 2019-05-02 Bioverativ Therapeutics Inc. Factor IX polypeptide formulations
CA2906864A1 (en) 2013-03-15 2014-09-18 Ampio Pharmaceuticals, Inc. Compositions for the mobilization, homing, expansion and differentiation of stem cells and methods of using the same
CN106913865A (en) 2013-04-18 2017-07-04 ้˜ฟๅฐ”่Žซ็”Ÿ็‰ฉ็ง‘ๆŠ€่‚กไปฝๆœ‰้™ๅ…ฌๅธ The method that disease and illness are treated using interleukin 10
CA2914837A1 (en) 2013-06-17 2014-12-24 Armo Biosciences, Inc. Method for assessing protein identity and stability
CN103408669B (en) 2013-08-01 2016-01-20 ๆฑŸ่‹ๆณฐๅบท็”Ÿ็‰ฉๅŒป่ฏๆœ‰้™ๅ…ฌๅธ GLP-1 analog fusion, and its production and use
WO2015031316A1 (en) 2013-08-30 2015-03-05 Armo Biosciences, Inc. Methods of using interleukin-10 for treating diseases and disorders
DK3063275T3 (en) 2013-10-31 2019-11-25 Resolve Therapeutics Llc THERAPEUTIC NUCLEASE ALBUMIN MERGERS AND PROCEDURES
US10208102B2 (en) 2013-11-01 2019-02-19 University Of Oslo Albumin variants and uses thereof
JP6660297B2 (en) 2013-11-11 2020-03-11 ใ‚ขใƒซใƒขใƒปใƒใ‚คใ‚ชใ‚ตใ‚คใ‚จใƒณใ‚ทใƒผใ‚บใƒปใ‚คใƒณใ‚ณใƒผใƒใƒฌใ‚คใƒ†ใƒƒใƒ‰ Methods of using interleukin-10 to treat diseases and disorders
WO2015077717A1 (en) 2013-11-25 2015-05-28 The Broad Institute Inc. Compositions and methods for diagnosing, evaluating and treating cancer by means of the dna methylation status
WO2015085147A1 (en) 2013-12-05 2015-06-11 The Broad Institute Inc. Polymorphic gene typing and somatic change detection using sequencing data
KR20160101073A (en) 2013-12-20 2016-08-24 ๋” ๋ธŒ๋กœ๋“œ ์ธ์Šคํ‹ฐํŠœํŠธ, ์ธ์ฝ”ํผ๋ ˆ์ดํ‹ฐ๋“œ Combination therapy with neoantigen vaccine
US10293043B2 (en) 2014-06-02 2019-05-21 Armo Biosciences, Inc. Methods of lowering serum cholesterol
JP6749312B2 (en) 2014-07-31 2020-09-02 ใ‚ขใƒ ใ‚ฒใƒณ ใƒชใ‚ตใƒผใƒ ๏ผˆใƒŸใƒฅใƒณใƒ˜ใƒณ๏ผ‰ ใ‚ฒใƒผใ‚จใƒ ใƒ™ใƒผใƒใƒผ๏ผก๏ผญ๏ผง๏ผฅ๏ผฎ ๏ผฒ๏ฝ…๏ฝ“๏ฝ…๏ฝ๏ฝ’๏ฝƒ๏ฝˆ๏ผˆ๏ผญ๏ฝ•๏ฝŽ๏ฝ‰๏ฝƒ๏ฝˆ๏ผ‰๏ผง๏ฝ๏ฝ‚๏ผจ Optimized interspecific bispecific single chain antibody constructs
US20170216403A1 (en) 2014-08-12 2017-08-03 Massachusetts Institute Of Technology Synergistic tumor treatment with il-2, a therapeutic antibody, and an immune checkpoint blocker
US10166273B2 (en) 2014-08-12 2019-01-01 The Board Of Trustees Of The Leland Stanford Junior University Synergistic tumor treatment with antibodies targeting PD-1, PD-L1 or CTLA4 and integrin-binding-Fc-fusion protein
CN106604915A (en) 2014-08-18 2017-04-26 ๅฎ‰็šฎๅฅฅๅˆถ่ฏ่‚กไปฝๆœ‰้™ๅ…ฌๅธ Treatment of joint conditions
EP3206713A4 (en) 2014-10-14 2018-06-27 Armo Biosciences, Inc. Interleukin-15 compositions and uses thereof
ES2941234T3 (en) 2014-10-22 2023-05-19 Armo Biosciences Inc Methods of using interleukin-10 for the treatment of diseases and disorders
US10993997B2 (en) 2014-12-19 2021-05-04 The Broad Institute, Inc. Methods for profiling the t cell repertoire
EP3234193B1 (en) 2014-12-19 2020-07-15 Massachusetts Institute of Technology Molecular biomarkers for cancer immunotherapy
WO2016126615A1 (en) 2015-02-03 2016-08-11 Armo Biosciences, Inc. Methods of using interleukin-10 for treating diseases and disorders
AU2016228806B2 (en) 2015-03-12 2020-10-22 Medimmune, Llc Method of purifying albumin-fusion proteins
JP2016179951A (en) * 2015-03-23 2016-10-13 ๅญฆๆ กๆณ•ไบบ ไธญๅคฎๅคงๅญฆ Transgenic animal serum albumin, hemoglobin-transgenic animal serum albumin complex, artificial plasma expander, and artificial oxygen carrier
FI3283524T3 (en) 2015-04-17 2023-05-05 Amgen Res Munich Gmbh Bispecific antibody constructs for cdh3 and cd3
CN116196401A (en) 2015-05-20 2023-06-02 ๅšๅพท็ ”็ฉถๆ‰€ Consensus neoantigens
JP7121496B2 (en) 2015-05-28 2022-08-18 ใ‚ขใƒซใƒขใƒปใƒใ‚คใ‚ชใ‚ตใ‚คใ‚จใƒณใ‚ทใƒผใ‚บใƒปใ‚คใƒณใ‚ณใƒผใƒใƒฌใ‚คใƒ†ใƒƒใƒ‰ Pegylated interleukin-10 for use in cancer therapy
US10781246B2 (en) 2015-06-05 2020-09-22 New York University Compositions and methods for anti-staphylococcal biologic agents
WO2016209969A1 (en) 2015-06-22 2016-12-29 Ampio Pharmaceuticals, Inc. Use of low molecular weight fractions of human serum albumin in treating diseases
TWI829617B (en) 2015-07-31 2024-01-21 ๅพทๅ•†ๅฎ‰็พŽๅŸบ็ ”็ฉถ๏ผˆๆ…•ๅฐผ้ป‘๏ผ‰ๅ…ฌๅธ Antibody constructs for flt3 and cd3
TWI717375B (en) 2015-07-31 2021-02-01 ๅพทๅ•†ๅฎ‰็พŽๅŸบ็ ”็ฉถ๏ผˆๆ…•ๅฐผ้ป‘๏ผ‰ๅ…ฌๅธ Antibody constructs for cd70 and cd3
TW202346349A (en) 2015-07-31 2023-12-01 ๅพทๅ•†ๅฎ‰็พŽๅŸบ็ ”็ฉถ๏ผˆๆ…•ๅฐผ้ป‘๏ผ‰ๅ…ฌๅธ Antibody constructs for dll3 and cd3
TWI796283B (en) 2015-07-31 2023-03-21 ๅพทๅ•†ๅฎ‰็พŽๅŸบ็ ”็ฉถ๏ผˆๆ…•ๅฐผ้ป‘๏ผ‰ๅ…ฌๅธ Antibody constructs for msln and cd3
TWI744242B (en) 2015-07-31 2021-11-01 ๅพทๅ•†ๅฎ‰็พŽๅŸบ็ ”็ฉถ๏ผˆๆ…•ๅฐผ้ป‘๏ผ‰ๅ…ฌๅธ Antibody constructs for egfrviii and cd3
AU2016312510A1 (en) 2015-08-25 2018-03-08 Armo Biosciences, Inc. Methods of using Interleukin-10 for treating diseases and disorders
CA3000742A1 (en) 2015-10-02 2017-04-06 Silver Creek Pharmaceuticals, Inc. Bi-specific therapeutic proteins for tissue repair
BR112018012851A2 (en) 2015-12-22 2018-12-26 Albumedix Ltd improved protein expression strains
WO2017112549A1 (en) 2015-12-23 2017-06-29 Biogen Ma Inc. Dkk2 cysteine rich domain 2 containing proteins and uses thereof
SG10201912498YA (en) 2016-01-07 2020-02-27 CSL Behring Lengnau AG Mutated truncated von willebrand factor
EA039859B1 (en) 2016-02-03 2022-03-21 ะญะผะดะถะตะฝ ะ ะธัะตั€ั‡ (ะœัŽะฝะธะบ) ะ“ะผะฑั… Bispecific antibody constructs binding egfrviii and cd3
WO2017184590A1 (en) 2016-04-18 2017-10-26 The Broad Institute Inc. Improved hla epitope prediction
US11110122B2 (en) 2016-06-03 2021-09-07 New York University Methods and reagents for modulating macrophage phenotype
KR102573478B1 (en) 2016-10-04 2023-09-04 ์•Œ๋ถ€๋ฉ”๋”•์Šค ๋ฆฌ๋ฏธํ‹ฐ๋“œ Uses of Recombinant Yeast-Derived Serum Albumin
WO2018082758A1 (en) 2016-11-04 2018-05-11 Aarhus Universitet Identification and treatment of tumors characterized by an overexpression of the neonatal fc receptor
WO2018096396A1 (en) * 2016-11-22 2018-05-31 University Of Oslo Albumin variants and uses thereof
US10268924B2 (en) * 2016-12-05 2019-04-23 Sap Se Systems and methods for integrated cargo inspection
JP2020502203A (en) 2016-12-21 2020-01-23 ใƒ—ใƒญใƒกใƒ†ใ‚ฃใƒƒใ‚ฏใƒปใƒ•ใ‚กใƒผใƒžใƒปใ‚จใ‚นใ‚จใƒ ใƒ†ใ‚ฃใƒปใƒชใƒŸใƒ†ใƒƒใƒ‰๏ผฐ๏ฝ’๏ฝ๏ฝ๏ฝ…๏ฝ”๏ฝ‰๏ฝƒ ๏ผฐ๏ฝˆ๏ฝ๏ฝ’๏ฝ๏ฝ ๏ผณ๏ฝ๏ฝ” ๏ผฌ๏ฝ‰๏ฝ๏ฝ‰๏ฝ”๏ฝ…๏ฝ„ Methods and compositions for preventing or minimizing epithelial-mesenchymal transition
EP3574116A1 (en) 2017-01-24 2019-12-04 The Broad Institute, Inc. Compositions and methods for detecting a mutant variant of a polynucleotide
KR20230119735A (en) 2017-02-12 2023-08-16 ๋ฐ”์ด์˜ค์—”ํ…Œํฌ ์œ ์—์Šค ์ธํฌ. Hla-based methods and compositions and uses thereof
EP3642343B1 (en) 2017-06-20 2021-12-08 Albumedix Ltd. Improved protein expression strains
KR20200018690A (en) 2017-06-22 2020-02-19 ์ฒด์—์Šค์—˜ ๋ฒ ๋ง ๋ ๋‚˜์šฐ ์•„๊ฒŒ Modulation of FVIII Immunogenicity by Cleaved VWF
CN111164100B (en) 2017-08-03 2024-03-12 ็พŽๅ›ฝๅฎ‰่ฟ›ๅ…ฌๅธ Interleukin-21 muteins and methods of treatment
WO2019036605A2 (en) 2017-08-17 2019-02-21 Massachusetts Institute Of Technology Multiple specificity binders of cxc chemokines and uses thereof
UY37870A (en) 2017-09-08 2019-03-29 Amgen Inc KRAS G12C INHIBITORS AND METHODS TO USE THEM FIELD OF THE INVENTION
KR20200067158A (en) * 2017-10-18 2020-06-11 ์”จ์—์Šค์—˜ ๋ฆฌ๋ฏธํ‹ฐ๋“œ Human serum albumin variants and uses thereof
PL3717011T3 (en) 2017-11-29 2023-03-27 Csl Limited Method of treating or preventing ischemia-reperfusion injury
TW201930344A (en) 2018-01-12 2019-08-01 ็พŽๅ•†ๅฎ‰้€ฒๅ…ฌๅธ Anti-PD-1 antibodies and methods of treatment
CN112118859A (en) 2018-05-16 2020-12-22 ๆฐ็‰นๆœ‰้™ๅ…ฌๅธ Type 1 soluble complement receptor variants and uses thereof
MA53330A (en) 2018-08-03 2021-06-09 Amgen Inc ANTIBODY CONSTRUCTIONS FOR CLDN18.2 AND CD3
EP3847246A1 (en) 2018-09-06 2021-07-14 Bavarian Nordic A/S Storage improved poxvirus compositions
US20210382068A1 (en) 2018-10-02 2021-12-09 Dana-Farber Cancer Institute, Inc. Hla single allele lines
JP7047932B2 (en) * 2018-10-25 2022-04-06 ๆ—ฅๆœฌ่ฃฝ้‰„ๆ ชๅผไผš็คพ A coating liquid for forming an insulating film for grain-oriented electrical steel sheets, a method for manufacturing grain-oriented electrical steel sheets, and a method for manufacturing grain-oriented electrical steel sheets.
EP3873934A2 (en) 2018-10-29 2021-09-08 Biogen MA Inc. Humanized and stabilized fc5 variants for enhancement of blood brain barrier transport
WO2020131586A2 (en) 2018-12-17 2020-06-25 The Broad Institute, Inc. Methods for identifying neoantigens
WO2020132586A1 (en) 2018-12-21 2020-06-25 Neon Therapeutics, Inc. Method and systems for prediction of hla class ii-specific epitopes and characterization of cd4+ t cells
SG11202108857SA (en) 2019-03-18 2021-09-29 Biontech Cell & Gene Therapies Gmbh Lnterleukin-2 receptor (il2r) and interleukin-2 (il2) variants for specific activation of immune effector cells
WO2020200481A1 (en) 2019-04-05 2020-10-08 Biontech Rna Pharmaceuticals Gmbh Treatment involving interleukin-2 (il2) and interferon (ifn)
WO2021044361A1 (en) 2019-09-06 2021-03-11 Novartis Ag Therapeutic fusion proteins
EP3819007A1 (en) 2019-11-11 2021-05-12 Amgen Research (Munich) GmbH Dosing regimen for anti-bcma agents
EP4069200A1 (en) 2019-12-04 2022-10-12 Albumedix Ltd Methods and compositions produced thereby
US20230093169A1 (en) 2020-01-22 2023-03-23 Amgen Research (Munch) Gmbh Combinations of antibody constructs and inhibitors of cytokine release syndrome and uses thereof
EP4118113A1 (en) 2020-03-12 2023-01-18 Amgen Inc. Method for treatment and prophylaxis of crs in patients comprising a combination of bispecifc antibodies binding to cds x cancer cell and tnfalpha or il-6 inhibitor
IL296250A (en) 2020-03-12 2022-11-01 Bavarian Nordic As Compositions improving poxvirus stability
JP2023518935A (en) 2020-03-16 2023-05-09 ใƒใ‚คใ‚ชใ‚จใƒŒใƒ†ใƒƒใ‚ฏ ใ‚ปใƒซ ใ‚ขใƒณใƒ‰ ใ‚ธใƒผใƒณ ใ‚ปใƒฉใƒ”ใƒผใ‚บ ใ‚ฒใƒผใ‚จใƒ ใƒ™ใƒผใƒใƒผ Antigen-specific T-cell receptors and T-cell epitopes
GB202004514D0 (en) 2020-03-27 2020-05-13 Inst De Medicina Molecular Joaeo Lobo Antunes Treatment of Immunosuppressive Cancer
WO2021236638A1 (en) 2020-05-19 2021-11-25 Amgen Inc. Mageb2 binding constructs
US20230398147A1 (en) 2020-09-16 2023-12-14 Amgen Inc. Methods for administering therapeutic doses of bispecific t-cell engaging molecules for the treatment of cancer
CR20230235A (en) 2020-11-06 2023-10-05 Amgen Res Munich Gmbh POLYPEPTIDE CONSTRUCTIONS THAT SELECTIVELY BIND CLDN6 AND CD3
TW202233678A (en) 2020-11-06 2022-09-01 ๅพทๅ•†ๅฎ‰็พŽๅŸบ็ ”็ฉถ๏ผˆๆ…•ๅฐผ้ป‘๏ผ‰ๅ…ฌๅธ Polypeptides with enhanced clipping profile
WO2022096698A1 (en) 2020-11-06 2022-05-12 Amgen Inc. Polypeptide constructs binding to cd3
TW202233682A (en) 2020-11-10 2022-09-01 ็พŽๅ•†ๅฎ‰้€ฒๅ…ฌๅธ Methods for administering a bcmaxcd3 binding molecule
KR20230142704A (en) 2020-12-14 2023-10-11 ๋ฐ”์ด์˜ค์—”ํ…Œํฌ ์œ ์—์Šค ์ธํฌ. Tissue-specific antigens for cancer immunotherapy
WO2024015892A1 (en) 2022-07-13 2024-01-18 The Broad Institute, Inc. Hla-ii immunopeptidome methods and systems for antigen discovery
WO2024077044A1 (en) 2022-10-05 2024-04-11 Amgen Inc. Combination therapies comprising t-cell redirecting therapies and agonistic anti-il-2r antibodies or fragments thereof
WO2024077256A1 (en) 2022-10-07 2024-04-11 The General Hospital Corporation Methods and compositions for high-throughput discovery ofpeptide-mhc targeting binding proteins

Family Cites Families (166)

* Cited by examiner, โ€  Cited by third party
Publication number Priority date Publication date Assignee Title
US5625041A (en) 1990-09-12 1997-04-29 Delta Biotechnology Limited Purification of proteins
GB9019919D0 (en) 1990-09-12 1990-10-24 Delta Biotechnology Ltd Purification of proteins
US2714586A (en) 1951-06-25 1955-08-02 Phillips Petroleum Co Washing urea and thiourea containing adducts
US4302386A (en) 1978-08-25 1981-11-24 The Ohio State University Antigenic modification of polypeptides
US4741900A (en) 1982-11-16 1988-05-03 Cytogen Corporation Antibody-metal ion complexes
US4757006A (en) 1983-10-28 1988-07-12 Genetics Institute, Inc. Human factor VIII:C gene and recombinant methods for production
JPS60169498A (en) 1984-02-10 1985-09-02 Kyowa Hakko Kogyo Co Ltd Adult t cell leukemic viral antigen peptide derivative
ATE72838T1 (en) 1985-04-12 1992-03-15 Genetics Inst NEW PROCOAGULATION PROTEINS.
GR860984B (en) 1985-04-17 1986-08-18 Zymogenetics Inc Expression of factor vii and ix activities in mammalian cells
ES2052712T3 (en) 1987-04-09 1994-07-16 Delta Biotechnology Ltd YEAST VECTOR.
ES2076939T3 (en) 1987-08-28 1995-11-16 Novo Nordisk As RECOMBINANT LUMPY OF HUMICOLA AND PROCEDURE FOR THE PRODUCTION OF RECOMBINANT LIPAS OF HUMICOLA.
GB8725529D0 (en) 1987-10-30 1987-12-02 Delta Biotechnology Ltd Polypeptides
IL88326A (en) 1987-11-18 1993-03-15 Gist Brocades Nv Purification of serum albumin
US5075222A (en) 1988-05-27 1991-12-24 Synergen, Inc. Interleukin-1 inhibitors
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5766883A (en) 1989-04-29 1998-06-16 Delta Biotechnology Limited Polypeptides
GB8909916D0 (en) 1989-04-29 1989-06-14 Delta Biotechnology Ltd Polypeptides
FR2650598B1 (en) 1989-08-03 1994-06-03 Rhone Poulenc Sante DERIVATIVES OF ALBUMIN WITH THERAPEUTIC FUNCTION
US5073627A (en) 1989-08-22 1991-12-17 Immunex Corporation Fusion proteins comprising GM-CSF and IL-3
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
GB8927722D0 (en) 1989-12-07 1990-02-07 British Bio Technology Proteins and nucleic acids
DE4000939A1 (en) 1990-01-15 1991-07-18 Brem Gottfried Prof Dr Dr METHOD FOR OBTAINING ANTIBODIES
JP3230091B2 (en) 1990-06-25 2001-11-19 ใ‚ฆใ‚งใƒซใƒ•ใ‚กใ‚คใƒ‰ๆ ชๅผไผš็คพ Method for suppressing coloration of human serum albumin
US5698426A (en) 1990-09-28 1997-12-16 Ixsys, Incorporated Surface expression libraries of heteromeric receptors
IL99552A0 (en) 1990-09-28 1992-08-18 Ixsys Inc Compositions containing procaryotic cells,a kit for the preparation of vectors useful for the coexpression of two or more dna sequences and methods for the use thereof
CA2058820C (en) 1991-04-25 2003-07-15 Kotikanyad Sreekrishna Expression cassettes and vectors for the secretion of human serum albumin in pichia pastoris cells
US5264586A (en) 1991-07-17 1993-11-23 The Scripps Research Institute Analogs of calicheamicin gamma1I, method of making and using the same
FR2686899B1 (en) 1992-01-31 1995-09-01 Rhone Poulenc Rorer Sa NOVEL BIOLOGICALLY ACTIVE POLYPEPTIDES, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
US5663060A (en) 1992-04-07 1997-09-02 Emory University Hybrid human/animal factor VIII
ZA932522B (en) 1992-04-10 1993-12-20 Res Dev Foundation Immunotoxins directed against c-erbB-2(HER/neu) related surface antigens
DE4226971C2 (en) 1992-08-14 1997-01-16 Widmar Prof Dr Tanner Modified fungal cells and processes for the production of recombinant products
US5728553A (en) 1992-09-23 1998-03-17 Delta Biotechnology Limited High purity albumin and method of producing
JP3095175B2 (en) 1992-11-13 2000-10-03 ใ‚ขใ‚คใƒ‡ใƒƒใ‚ฏ ใƒ•ใ‚กใƒผใƒžใ‚ทใƒฅใƒผใƒ†ใ‚ฃใ‚ซใƒซใ‚บ ใ‚ณใƒผใƒใƒฌใ‚คใ‚ทใƒงใƒณ Therapeutic use of chimeric and radiolabeled antibodies against human B lymphocyte restricted differentiation antigen for the treatment of B cell lymphoma
CA2167580A1 (en) 1993-07-22 1995-02-02 Howard Y. Chen Expression of human interleukin-1.beta. in a transgenic animal
DE4343591A1 (en) 1993-12-21 1995-06-22 Evotec Biosystems Gmbh Process for the evolutionary design and synthesis of functional polymers based on shape elements and shape codes
US6811773B1 (en) 1993-12-22 2004-11-02 Human Genome Sciences, Inc. Human monocyte colony inhibitory factor (M-CIF) polypeptides
US5605793A (en) 1994-02-17 1997-02-25 Affymax Technologies N.V. Methods for in vitro recombination
GB9404270D0 (en) * 1994-03-05 1994-04-20 Delta Biotechnology Ltd Yeast strains and modified albumins
US5773001A (en) 1994-06-03 1998-06-30 American Cyanamid Company Conjugates of methyltrithio antitumor agents and intermediates for their synthesis
US7597886B2 (en) 1994-11-07 2009-10-06 Human Genome Sciences, Inc. Tumor necrosis factor-gamma
US5714586A (en) 1995-06-07 1998-02-03 American Cyanamid Company Methods for the preparation of monomeric calicheamicin derivative/carrier conjugates
US5712374A (en) 1995-06-07 1998-01-27 American Cyanamid Company Method for the preparation of substantiallly monomeric calicheamicin derivative/carrier conjugates
US5716808A (en) 1995-11-09 1998-02-10 Zymogenetics, Inc. Genetic engineering of pichia methanolica
US5955349A (en) 1996-08-26 1999-09-21 Zymogenetics, Inc. Compositions and methods for producing heterologous polypeptides in Pichia methanolica
GB9526733D0 (en) 1995-12-30 1996-02-28 Delta Biotechnology Ltd Fusion proteins
US6509313B1 (en) 1996-02-28 2003-01-21 Cornell Research Foundation, Inc. Stimulation of immune response with low doses of cytokines
CN1230997A (en) 1996-07-17 1999-10-06 ๆดฅ่Žซๅ‰ๅฐผ่’‚ๅ…‹ๆ–ฏๅ…ฌๅธ Transformation of pichia methanolica
US5736383A (en) 1996-08-26 1998-04-07 Zymogenetics, Inc. Preparation of Pichia methanolica auxotrophic mutants
US6274305B1 (en) 1996-12-19 2001-08-14 Tufts University Inhibiting proliferation of cancer cells
US6605699B1 (en) 1997-01-21 2003-08-12 Human Genome Sciences, Inc. Galectin-11 polypeptides
US7196164B2 (en) 1997-07-08 2007-03-27 Human Genome Sciences, Inc. Secreted protein HHTLF25
US7053190B2 (en) 1997-03-07 2006-05-30 Human Genome Sciences, Inc. Secreted protein HRGDF73
US6506569B1 (en) 1997-05-30 2003-01-14 Human Genome Sciences, Inc. Antibodies to human tumor necrosis factor receptor TR10
US5948609A (en) 1997-12-03 1999-09-07 Carter; Daniel C. Oxygen-transporting albumin-based blood replacement composition and blood volume expander
EP1030931A4 (en) 1998-08-06 2004-05-26 Syntron Biores Inc Uric acid assay device with stabilized uricase reagent composition
CA2356542A1 (en) 1998-12-23 2000-07-06 Human Genome Sciences, Inc. Peptidoglycan recognition proteins
GB9902000D0 (en) 1999-01-30 1999-03-17 Delta Biotechnology Ltd Process
DE60043021D1 (en) 1999-05-17 2009-11-05 Conjuchem Biotechnologies Inc Long-acting peptide inhibitors of viral fusion with body cells in viral infections
EP1178786A4 (en) 1999-05-21 2006-03-01 American Bioscience Inc Protein stabilized pharmacologically active agents, methods for the preparation thereof and methods for the use thereof
DK1266006T3 (en) 2000-03-22 2006-01-16 Octagene Gmbh Preparation of Recombinant Muteins of Blood Coagulation Factor VIII in Human Cell Lines
EP1274720A4 (en) 2000-04-12 2004-08-18 Human Genome Sciences Inc Albumin fusion proteins
AU2001288301A1 (en) 2000-08-18 2002-03-04 Human Genome Sciences, Inc. Binding polypeptides and methods based thereon
JP2005500806A (en) 2000-09-15 2005-01-13 ใ‚ณใƒผใƒชใƒผ ใƒ•ใ‚กใƒผใƒžใ‚ทใƒฅใƒผใƒ†ใ‚ฃใ‚ซใƒซ ใ‚ฒใƒผใ‚จใƒ ใƒ™ใƒผใƒใƒผ Process for high-throughput screening of immune agonist / immunoantagonists based on CpG
WO2002055110A2 (en) 2000-10-25 2002-07-18 Genzyme Corp Methods for treating blood coagulation disorders
US6992234B2 (en) 2000-11-06 2006-01-31 The Jackson Laboratory FcRn-based therapeutics for the treatment of auto-immune disorders
PL393178A1 (en) 2000-12-07 2011-02-14 Eli Lilly And Company Heterogeneous fusion protein, pharmaceutical composition for the treatment of diabetic patients with insulin-independent diabetes and pharmaceutical composition for the treatment of obese patients
US7175988B2 (en) 2001-02-09 2007-02-13 Human Genome Sciences, Inc. Human G-protein Chemokine Receptor (CCR5) HDGNR10
US7507413B2 (en) 2001-04-12 2009-03-24 Human Genome Sciences, Inc. Albumin fusion proteins
ATE470676T1 (en) 2001-04-13 2010-06-15 Human Genome Sciences Inc ANTI-VEGF-2 ANTIBODIES
AUPR446701A0 (en) 2001-04-18 2001-05-17 Gene Stream Pty Ltd Transgenic mammals for pharmacological and toxicological studies
WO2002102830A1 (en) 2001-06-15 2002-12-27 New Century Pharmaceuticals, Inc. Human albumin animal models for drug evaluation, toxicology and immunogenicity studies
AUPR622301A0 (en) 2001-07-09 2001-08-02 Novapharm Research (Australia) Pty Ltd Infection control system
ATE375388T1 (en) 2001-07-27 2007-10-15 Us Gov Health & Human Serv SYSTEMS FOR SITE-DIRECTED IN VIVO MUTAGENesis USING OLIGONUCLEOTIDES
CN1405182A (en) * 2001-08-10 2003-03-26 ไธญๅ›ฝไบบๆฐ‘่งฃๆ”พๅ†›ๅ†›ไบ‹ๅŒปๅญฆ็ง‘ๅญฆ้™ข็”Ÿ็‰ฉๅทฅ็จ‹็ ”็ฉถๆ‰€ Serum albumin and granulocyte colony stimulating factor fusion protein
CA2484556A1 (en) 2001-12-21 2003-07-24 Human Genome Sciences, Inc. Albumin fusion proteins
CA2841097A1 (en) 2001-12-21 2003-07-24 Human Genome Sciences, Inc. Albumin and g-csf fusion proteins
US20080167238A1 (en) 2001-12-21 2008-07-10 Human Genome Sciences, Inc. Albumin Fusion Proteins
NZ579419A (en) 2002-02-07 2011-02-25 Dyax Corp Albumin-fused kunitz domain peptides
US20080108560A1 (en) 2002-03-05 2008-05-08 Eli Lilly And Company Heterologous G-Csf Fusion Proteins
WO2003102136A2 (en) 2002-05-30 2003-12-11 Human Genome Sciences, Inc. Antibodies that specifically bind to neurokinin b
CN1241946C (en) 2002-07-01 2006-02-15 ็พŽๅ›ฝ็ฆๆบ้›†ๅ›ข Human serum albumins recombined merge protein having hyperplasia stimulation function to multiple cells
GB0217347D0 (en) * 2002-07-26 2002-09-04 Univ Edinburgh Novel albumins
CA2492457A1 (en) 2002-08-02 2004-02-12 Craig A. Rosen Antibodies against c3a receptor
WO2004058044A2 (en) 2002-11-19 2004-07-15 Drg International, Inc. Diagnostic method for diseases by screening for hepcidin in human or animal tissues, blood or body fluids and therapeutic uses therefor
CA2513213C (en) 2003-01-22 2013-07-30 Human Genome Sciences, Inc. Albumin fusion proteins
ATE411819T1 (en) 2003-02-17 2008-11-15 Upperton Ltd CONJUGATES FOR MEDICAL IMAGING COMPRISING CARRIERS, TARGETING MOTIONS AND CONTRAST AGENTS
GB0305989D0 (en) 2003-03-15 2003-04-23 Delta Biotechnology Ltd Agent
WO2004082640A2 (en) 2003-03-19 2004-09-30 New Century Pharmaceuticals, Inc. Human serum albumin conjugates with therapeutic compounds
US20070166711A1 (en) 2003-04-15 2007-07-19 Samuels Mark E Juvenile hemochromatosis gene (hfe2a), expression products and uses thereof
US20050079546A1 (en) 2003-05-01 2005-04-14 Dasa Lipovsek Serum albumin scaffold-based proteins and uses thereof
WO2005007121A2 (en) 2003-07-18 2005-01-27 Massachusetts Institute Of Technology Mutant interleukin-2(il-2) polypeptides
US6949961B2 (en) 2003-10-06 2005-09-27 Semiconductor Components Industries, L.L.C. Power switch structure with low RDSon and low current limit
US20080308744A1 (en) 2003-11-18 2008-12-18 Beth Israel Deaconess Medical Center Serum Albumin Conjugated to Fluorescent Substances for Imaging
GB0329681D0 (en) 2003-12-23 2004-01-28 Delta Biotechnology Ltd Gene expression technique
GB0329722D0 (en) 2003-12-23 2004-01-28 Delta Biotechnology Ltd Modified plasmid and use thereof
JP4649954B2 (en) 2003-12-26 2011-03-16 ใƒ‹ใƒ—ใƒญๆ ชๅผไผš็คพ Albumin with enhanced antibacterial activity
US7166577B2 (en) 2003-12-26 2007-01-23 Nipro Corporation Albumin having enhanced antimicrobial activity
KR100671005B1 (en) 2004-01-15 2007-01-18 ๊ณ ๋ ค๋Œ€ํ•™๊ต ์‚ฐํ•™ํ˜‘๋ ฅ๋‹จ Biomarker proteins for diagnosing the exposure to PAH
RU2369404C2 (en) * 2004-02-09 2009-10-10 ะฅัŒัŽะผะฐะฝ ะ”ะถะตะฝะพะผ ะกะฐะนะตะฝัะธะท, ะ˜ะฝะบ. Fused proteins of albumine
BRPI0507026A (en) 2004-02-09 2007-04-17 Human Genome Sciences Inc albumin fusion proteins
JP4492156B2 (en) 2004-03-03 2010-06-30 ใƒ‹ใƒ—ใƒญๆ ชๅผไผš็คพ Protein containing serum albumin domain
US20060018859A1 (en) 2004-07-16 2006-01-26 Carter Daniel C Modified human serum albumin with reduced or eliminated affinity to chemical or biological contaminants at Cys 34
CA2575714A1 (en) 2004-08-06 2006-02-09 Akihiro Meta Yeast promoter
US20060051859A1 (en) * 2004-09-09 2006-03-09 Yan Fu Long acting human interferon analogs
JP4948413B2 (en) 2004-09-23 2012-06-06 ใ‚ธใ‚งใƒใƒณใƒ†ใƒƒใ‚ฏ๏ผŒ ใ‚คใƒณใ‚ณใƒผใƒใƒฌใ‚คใƒ†ใƒƒใƒ‰ Cysteine engineered antibodies and conjugates
US20090280534A1 (en) 2004-12-22 2009-11-12 Novozymes A/S Recombinant Production of Serum Albumin
EP2330200B1 (en) 2004-12-23 2017-04-05 Albumedix A/S Gene expression technique
WO2006073195A1 (en) 2005-01-07 2006-07-13 Biomarker Science Co., Ltd Method for predicting or diagnosing diabetes and kit for predicting or diagnosing diabetes
US20060178301A1 (en) 2005-02-04 2006-08-10 Mathias Jurs Albumin-fused ciliary neurotrophic factor
EP1879920A2 (en) 2005-04-29 2008-01-23 The Jackson Laboratory Fcrn antibodies and uses thereof
GB0512707D0 (en) 2005-06-22 2005-07-27 Delta Biotechnology Ltd Gene expression technique
WO2007021494A2 (en) * 2005-08-12 2007-02-22 Human Genome Sciences, Inc. Albumin fusion proteins
CN101287750A (en) * 2005-08-12 2008-10-15 ไบบ็ฑปๅŸบๅ› ็ง‘ๅญฆๅ…ฌๅธ Albumin fusion proteins
CA2562249A1 (en) * 2005-10-20 2007-04-20 University Of Ottawa Heart Institute Anf analogue
CA2634495A1 (en) 2005-12-22 2007-06-28 Conjuchem Biotechnologies Inc. Process for the production of preformed conjugates of albumin and a therapeutic agent
EP1816201A1 (en) 2006-02-06 2007-08-08 CSL Behring GmbH Modified coagulation factor VIIa with extended half-life
WO2007112940A2 (en) 2006-03-31 2007-10-11 Ablynx N.V. Albumin-derived amino acid sequence, use thereof for increasing the half-life of therapeutic proteins and of other therapeutic compounds and entities, and constructs comprising the same
EP2046367A4 (en) 2006-06-07 2009-11-11 Human Genome Sciences Inc Albumin fusion proteins
DK2256135T3 (en) 2006-06-14 2019-06-11 Csl Behring Gmbh Proteolytically cleavable fusion protein comprising a blood coagulation factor
CA2657277C (en) 2006-07-13 2015-11-24 Upperton Limited Process for preparing particles of proteinaceous material
JP4983148B2 (en) 2006-08-18 2012-07-25 ใƒ‹ใƒ—ใƒญๆ ชๅผไผš็คพ Glycan-containing albumin, method for producing the same, and use thereof
AU2007292903B2 (en) 2006-09-08 2012-03-29 Ambrx, Inc. Modified human plasma polypeptide or Fc scaffolds and their uses
US20100129846A1 (en) 2006-12-07 2010-05-27 Power3 Medical Products, Inc. Isoform specificities of blood serum proteins and their use as differentially expressed protein biomarkers for diagnosis of breast cancer
JP2010535484A (en) 2007-08-08 2010-11-25 ใƒŽใƒœใ‚ถใ‚คใƒ ใ‚น ใƒใ‚คใ‚ชใƒ•ใ‚กใƒผใƒž ใƒ‡ใƒผใ‚ณใƒผ ใ‚ขใ‚ฏใƒ†ใ‚ฃใƒผใ‚ผใƒซใ‚นใ‚ซใƒ– Transferrin mutants and complexes
WO2009061853A2 (en) 2007-11-05 2009-05-14 Massachusetts Institute Of Technology Mutant interleukin-2 (il-2) polypeptides
CA2611540C (en) 2007-11-09 2017-05-30 Nipro Corporation Sugar chain-containing albumin as a drug carrier to the liver
MX343594B (en) 2007-12-21 2016-11-11 Medimmune Ltd BINDING MEMBERS FOR INTERLEUKIN-4 RECEPTOR ALPHA (IL-4Ra) - 173.
CA2721093A1 (en) 2008-04-11 2009-10-15 Merrimack Pharmaceuticals, Inc. Human serum albumin linkers and conjugates thereof
US20110112040A1 (en) 2008-04-28 2011-05-12 President And Fellows Of Harvard College Supercharged proteins for cell penetration
JP2011528804A (en) 2008-07-18 2011-11-24 ใ‚ชใƒฉใ‚ธใ‚งใƒ‹ใƒƒใ‚ฏใ‚น๏ผŒใ‚คใƒณใ‚ณใƒผใƒใƒฌใ‚คใƒ†ใƒƒใƒ‰ Composition for the detection and treatment of colorectal cancer
CN102282168A (en) * 2008-11-18 2011-12-14 ๆข…้‡Œ้บฆๅ…‹ๅˆถ่ฏ่‚กไปฝๆœ‰้™ๅ…ฌๅธ Human serum albumin linkers and conjugates thereof
AU2009322126B2 (en) 2008-12-05 2013-06-20 Abraxis Bioscience, Llc Albumin binding peptide-mediated disease targeting
CN102307905B (en) 2008-12-10 2015-11-25 ๆ–ฏๅ…‹ๅˆฉๆ™ฎๆ–ฏ็ ”็ฉถ้™ข Chemical reactivity alpha-non-natural amino acid is utilized to produce carrier-peptide conjugate
DK2396347T3 (en) * 2009-02-11 2017-07-24 Albumedix As ALBUMIN VARIANTS AND CONJUGATES
CN102458442B (en) 2009-04-08 2016-06-29 ๅŠ ๅˆฉ็ฆๅฐผไบšๅคงๅญฆ่‘ฃไบ‹ไผš There is people's albumen support of controlled serum pharmacokinetic
EP2424877A4 (en) 2009-04-28 2013-01-02 Harvard College Supercharged proteins for cell penetration
US8232067B2 (en) 2009-05-29 2012-07-31 Brigham & Women's Hospital, Inc. Disrupting FCRN-albumin interactions
ES2548030T3 (en) 2009-06-01 2015-10-13 Medimmune, Llc Molecules with prolonged half-lives and uses thereof
KR101286721B1 (en) 2009-06-05 2013-07-16 ํ•œ๊ตญ๊ณผํ•™๊ธฐ์ˆ ์—ฐ๊ตฌ์› Recombinant albumins fused with poly-cysteine peptide and the methods for preparing the same
EP2456454A4 (en) 2009-07-20 2013-03-20 Univ Nat Cheng Kung Polypeptides selective for av 3 integrin conjugated with a variant of human serum albumin (hsa) and pharmaceutical uses thereof
WO2011011797A2 (en) 2009-07-24 2011-01-27 The Board Of Trustees Of The Leland Stanford Junior University Cytokine compositions and methods of use thereof
CA2770620A1 (en) 2009-08-10 2011-02-17 Mark Smith Thiol protecting group
US20110172398A1 (en) 2009-10-02 2011-07-14 Boehringer Ingelheim International Gmbh Bispecific binding molecules for anti-angiogenesis therapy
KR20120093932A (en) 2009-10-10 2012-08-23 ์ผ๋ ˆ๋ธ ๋ฐ”์ด์˜คํ…Œ๋ผํ”ผํ‹ฑ์Šค, ์•„์ด์—”์”จ. Il-17 family cytokine compositions and uses
BR112012009450A2 (en) 2009-10-30 2017-05-23 Novozymes Biopharma Dk As albumin variants
WO2011079175A1 (en) 2009-12-23 2011-06-30 National Cheng Kung University Compositions and methods for the treatment of angiogenesis-related eye diseases
CN101875693B (en) 2010-01-22 2012-07-18 ๆˆ้ƒฝๆญฃ่ƒฝ็”Ÿ็‰ฉๆŠ€ๆœฏๆœ‰้™่ดฃไปปๅ…ฌๅธ Albumin variant having anti-angiogenesis activity and preparation method thereof
EP2536756B1 (en) 2010-02-16 2018-04-25 MedImmune, LLC Hsa-related compositions and methods of use
CN102939304B (en) * 2010-04-09 2017-04-19 ้˜ฟๅฐ”ๅธƒ้บฆ็‹„ๅ…‹ๆ–ฏๅ…ฌๅธ albumin derivatives and variants
WO2011146902A1 (en) 2010-05-21 2011-11-24 Merrimack Pharmaceuticals, Inc. Bi-specific fusion proteins
WO2011161127A1 (en) 2010-06-21 2011-12-29 Medimmune, Llc Protease variants of human neprilysin
JP2013538566A (en) 2010-08-13 2013-10-17 ใ‚ฐใƒฉใ‚ฏใ‚ฝใ‚นใƒŸใ‚นใ‚ฏใƒฉใ‚คใƒณใ€ใ‚คใƒณใƒ†ใƒฌใ‚ฏใƒใƒฅใ‚ขใƒซใ€ใƒ—ใƒญใƒ‘ใƒ†ใ‚ฃใƒผใ€ใƒ‡ใ‚ฃใƒ™ใƒญใƒƒใƒ—ใƒกใƒณใƒˆใ€ใƒชใƒŸใƒ†ใƒƒใƒ‰ Improved antiserum albumin binding variants
US20130225496A1 (en) 2010-11-01 2013-08-29 Novozymes Biopharma Dk A/S Albumin Variants
KR20140012094A (en) 2011-02-15 2014-01-29 ๋ฉ”๋””๋ฎจ ์—˜์—˜์”จ Hsa-related compositions and methods of use
US9045564B2 (en) 2011-02-15 2015-06-02 Medimmune, Llc HSA-related compositions and methods of use
EP2705051A1 (en) * 2011-05-05 2014-03-12 Novozymes Biopharma DK A/S Albumin variants
AU2012286048A1 (en) 2011-07-18 2014-02-20 Arts Biologics A/S Long acting luteinizing hormone (LH) compound
EP2780364A2 (en) 2011-11-18 2014-09-24 Eleven Biotherapeutics, Inc. Proteins with improved half-life and other properties
KR20140136934A (en) 2012-03-16 2014-12-01 ๋…ธ๋ณด์ž์ž„์Šค ๋ฐ”์ด์˜คํŒŒ๋งˆ ๋””์ผ€์ด ์—์ด/์—์Šค Albumin variants
WO2014005596A1 (en) 2012-07-03 2014-01-09 Aarhus Universitet Modified payload molecules and their interactions and uses
KR20150082422A (en) * 2012-11-08 2015-07-15 ๋…ธ๋ณด์ž์ž„์Šค ๋ฐ”์ด์˜คํŒŒ๋งˆ ๋””์ผ€์ด ์—์ด/์—์Šค Albumin variants
KR20150118123A (en) 2013-02-16 2015-10-21 ๋…ธ๋ณด์ž์ž„์Šค ๋ฐ”์ด์˜คํŒŒ๋งˆ ๋””์ผ€์ด ์—์ด/์—์Šค Pharmacokinetic animal model
WO2014179657A1 (en) 2013-05-03 2014-11-06 Eleven Biotherapeutics, Inc. Albumin variants binding to fcrn
WO2015036579A1 (en) 2013-09-13 2015-03-19 Novozymes Biopharma Dk A/S Albumin variants
US10208102B2 (en) * 2013-11-01 2019-02-19 University Of Oslo Albumin variants and uses thereof
CA2989966A1 (en) 2015-08-20 2017-02-23 Albumedix A/S Albumin variants and conjugates

Cited By (6)

* Cited by examiner, โ€  Cited by third party
Publication number Priority date Publication date Assignee Title
US11555061B2 (en) 2009-02-11 2023-01-17 Albumedix, Ltd Albumin variants and conjugates
US10711050B2 (en) 2011-11-18 2020-07-14 Albumedix Ltd Variant serum albumin with improved half-life and other properties
US10329340B2 (en) 2012-03-16 2019-06-25 Albumedix Ltd Albumin variants
US10501524B2 (en) 2012-11-08 2019-12-10 Albumedix Ltd Albumin variants
US10934341B2 (en) 2012-11-08 2021-03-02 Albumedix, Ltd. Albumin variants
US10633428B2 (en) 2015-08-20 2020-04-28 Albumedix Ltd Albumin variants and conjugates

Also Published As

Publication number Publication date
US10696732B2 (en) 2020-06-30
GB201209553D0 (en) 2012-07-11
EP3421491A2 (en) 2019-01-02
WO2011051489A3 (en) 2011-06-23
EP2493921B1 (en) 2018-09-26
US20140234311A1 (en) 2014-08-21
US8748380B2 (en) 2014-06-10
GB2488077A (en) 2012-08-15
CN102741280B (en) 2015-12-02
US20180105578A1 (en) 2018-04-19
US20180162925A1 (en) 2018-06-14
BR112012009450A2 (en) 2017-05-23
WO2011051489A2 (en) 2011-05-05
US20180105576A1 (en) 2018-04-19
AU2010311332B2 (en) 2015-04-23
US20160075757A1 (en) 2016-03-17
JP2018102300A (en) 2018-07-05
CN105567699A (en) 2016-05-11
US20120220530A1 (en) 2012-08-30
JP6703016B2 (en) 2020-06-03
JP6306628B2 (en) 2018-04-04
KR20120101403A (en) 2012-09-13
US20160075762A1 (en) 2016-03-17
US20160075756A1 (en) 2016-03-17
US20200385442A1 (en) 2020-12-10
RU2607374C2 (en) 2017-01-10
US20180105577A1 (en) 2018-04-19
AU2010311332A1 (en) 2012-04-26
US20160075761A1 (en) 2016-03-17
ES2700230T3 (en) 2019-02-14
IL218858B (en) 2018-10-31
US20160009787A1 (en) 2016-01-14
IL218858A0 (en) 2012-06-28
EP3421491A3 (en) 2019-03-27
EP2493921A2 (en) 2012-09-05
RU2012122173A (en) 2013-12-10
US20160075760A1 (en) 2016-03-17
MX2012004793A (en) 2012-07-20
US20160075758A1 (en) 2016-03-17
KR101874834B1 (en) 2018-07-05
US20160075763A1 (en) 2016-03-17
CA2776241A1 (en) 2011-05-05
JP2016165289A (en) 2016-09-15
US20180072792A1 (en) 2018-03-15
JP2013509170A (en) 2013-03-14
US20180222963A1 (en) 2018-08-09
US20160075759A1 (en) 2016-03-17
CN102741280A (en) 2012-10-17

Similar Documents

Publication Publication Date Title
US10696732B2 (en) Albumin variants
EP1427750B1 (en) Modified transferrin fusion proteins
EP1663278A2 (en) Epo mimetic peptides and fusion proteins
AU2002323501A1 (en) Modified transferrin fusion proteins
EP1951277A2 (en) Natriuretic peptide modified transferrin fusion proteins
JP2020100633A (en) Albumin variants
AU2015205821B2 (en) Albumin variants

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

AS Assignment

Owner name: ALBUMEDIX LTD, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NOVOZYMES BIOPHARMA DK A/S (BI-NAME ALBUMEDIX A/S);REEL/FRAME:046170/0127

Effective date: 20180412

AS Assignment

Owner name: NOVOZYMES BIOPHARMA UK LIMITED, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PLUMRIDGE, ANDREW;CAMERON, JASON;SLEEP, DARRELL;SIGNING DATES FROM 20101105 TO 20101116;REEL/FRAME:048319/0615

Owner name: UNIVERSITY OF OSLO, NORWAY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SANDLIE, INGER;ANDERSEN, JAN TERJE;REEL/FRAME:048319/0903

Effective date: 20101108

Owner name: NOVOZYMES A/S, DENMARK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:FRIIS, ESBEN PETER;REEL/FRAME:048320/0674

Effective date: 20111010

Owner name: NOVOZYMES BIOPHARMA DK A/S, DENMARK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:NOVOZYMES A/S;NOVOZYMES BIOPHARMA UK LIMITED;UNIVERSITY OF OSLO;SIGNING DATES FROM 20101029 TO 20111026;REEL/FRAME:048322/0949

Owner name: ALBUMEDIX A/S, DENMARK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NOVOZYMES BIOPHARMA DK A/S;REEL/FRAME:048329/0475

Effective date: 20160119

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STCF Information on status: patent grant

Free format text: PATENTED CASE

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1551); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 4